Optimizing Pharmacotherapy in Older Patients
Optimizing Pharmacotherapy in Older Patients
Antonio Cherubini
Arduino A. Mangoni
Denis O’Mahony
Mirko Petrovic Editors
Optimizing
Pharmacotherapy
in Older Patients
An Interdisciplinary Approach
Practical Issues in Geriatrics
Series Editor
Stefania Maggi, Aging Branch, CNR-Neuroscience Institute
Padua, Italy
This practically oriented series presents state of the art knowledge on the principal
diseases encountered in older persons and addresses all aspects of management,
including current multidisciplinary diagnostic and therapeutic approaches. It is
intended as an educational tool that will enhance the everyday clinical practice of
both young geriatricians and residents and also assist other specialists who deal
with aged patients. Each volume is designed to provide comprehensive information
on the topic that it covers, and whenever appropriate the text is complemented by
additional material of high educational and practical value, including informative
video-clips, standardized diagnostic flow charts and descriptive clinical cases.
Practical Issues in Geriatrics will be of value to the scientific and professional
community worldwide, improving understanding of the many clinical and social
issues in Geriatrics and assisting in the delivery of optimal clinical care.
Antonio Cherubini • Arduino A. Mangoni
Denis O’Mahony • Mirko Petrovic
Editors
Optimizing
Pharmacotherapy in Older
Patients
An Interdisciplinary Approach
Editors
Antonio Cherubini Arduino A. Mangoni
Geriatric Discipline of Clinical Pharmacology
Geriatric Admissions and Aging Research College of Medicine and Public Health,
Center, IRCCS INRCA Flinders University
Ancona, Italy Bedford Park, SA, Australia
This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Foreword
Aging seems to be a longer lasting pandemic than COVID. Older people enjoy liv-
ing happily despite a rapidly increasing disease burden, and this has to do with the
robust and expanding achievements of hygiene and medicine. This may or may not
include the most important option for treatment in almost all therapeutic areas,
which is the prescription of drugs. Medication can positively contribute to disease
prevention and mitigation, but due to inevitable side effects it could also be limiting
the enjoyment of life or even end it.
This topic of medication in older people thus is of paramount importance for this
steadily growing population part of our industrial societies. The new book on
Optimizing pharmacotherapy in older patients is one of the very few attempts to
condense our knowledge on this issue and comprehensively detail the dimensions,
implications, and technical accommodations for coping with this challenging aspect
of medicine. In its 29 chapters, the book deals with the epidemiology of “polyphar-
macy” (a widely used, but incorrect term which should rather be multimedication),
the detrimental impact of some pharmacodynamic and pharmacokinetic features of
the aging body on therapeutic outcomes and on instruments, practices, and tools to
optimize medication use in older people. Catchwords like “deprescribing” or
“potentially inappropriate medications” (PIM) are defined and discussed in the cur-
rent scientific context. Drug–drug, drug–nutrient, and drug–disease interactions are
being exemplified, issues of adherence and the roles of potential networking part-
ners such as pharmacists addressed in chapters by outstanding researchers in
the area.
The content of this first section on general issues is applied to geriatric syn-
dromes such as frailty or falls, and to special aspects of medications for frequent
diseases in older people (e.g., hypertension or heart failure) in the second main part
of the book.
It thus does not only provide the theoretical background but also practical,
disease-oriented solutions to medication-related problems in older people.
In its comprehensive approach, this book provides all relevant information for
enabling a better and more appropriate medication process in the geriatric popula-
tion. The book, thus, addresses physicians both in ambulatory and clinical care,
pharmacists, nursing staff, and all care providers involved in the complex process of
individualized medication in older people. Only few similar books have been
v
vi Foreword
published on this important issue to date, but none is comparably actual and
comprehensive.
We are convinced that this new book is instructive, helpful, and protective in this
context by not only showing the risks, but also the opportunities by the correct
application of drugs in older people. It clearly conveys that drugs may harm, but
also successfully treat older people if chosen correctly. The current FORTA-list con-
tains 41% of positively labelled drugs. Thus, the right drug for the right patient is
still a very good option for older people to live longer and/or better.
In the name of all those patients benefitting from the book, we sincerely thank
the editors and authors for the creation of this important work.
vii
viii Contents
10
Deprescribing: Evidence Base and Implementation������������������������������ 119
Denis Curtin and Denis O’Mahony
25
Optimizing Pharmacotherapy in Older Patients
with Depression or Anxiety ���������������������������������������������������������������������� 369
Sylvie Bonin-Guillaume
26
Nutritional Deficiency and Malnutrition ������������������������������������������������ 381
Eva Kiesswetter and Cornel C. Sieber
27
Pharmacotherapy of Insomnia in Older Adults�������������������������������������� 391
Mirko Petrovic
28
Optimizing Pharmacotherapy in Older Patients:
An Interdisciplinary Approach: Chronic Kidney Disease �������������������� 405
Andrea Corsonello, Antonello Rocca, Carmela Lo Russo,
and Luca Soraci
29
Palliation at End of Life���������������������������������������������������������������������������� 427
Joanne Droney, Phoebe Wright, and Dola Awoyemi
Index�������������������������������������������������������������������������������������������������������������������� 441
Part I
General Issues
The Impact of Ageing
on Pharmacokinetics 1
Arduino A. Mangoni and Elzbieta A. Jarmuzewska
1.1 Introduction
The age-associated accumulation and coexistence of disease states and the increas-
ingly proactive approach towards disease management by healthcare professionals
have led to an increased medication use in the older patient population [1, 2].
However, a concomitant increasing exposure to drugs that are potentially inappro-
priate has also been extensively reported, with the consequent risk of toxicity and its
clinical sequelae, for example, falls, cognitive impairment, hospitalisation and death
[3–8]. The predisposition of older people to the unintended effects of drug exposure
is largely due to specific alterations in body composition, organ function and homeo-
static capacity, both at the cellular and the system level, that may influence the
pharmacokinetics of several drugs [9]. Such alterations, in turn, lead to excessive
drug accumulation and an increased risk of drug–drug and drug–disease interac-
tions, particularly in the context of inappropriate prescribing and polypharmacy [9].
A better understanding of the main pharmacokinetic changes associated with
advancing age might play an important role in minimising the burden of inappropri-
ate polypharmacy. This burden is particularly high in frail patients, an ever-growing
subgroup characterised by reduced functional capacity and marked vulnerability to
adverse health outcomes [10].
A. A. Mangoni (*)
Discipline of Clinical Pharmacology, College of Medicine and Public Health,
Flinders University and Flinders Medical Centre, Adelaide, SA, Australia
Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local
Health Network, Adelaide, Australia
e-mail: [email protected]
E. A. Jarmuzewska
Department of Internal Medicine, Polyclinic IRCCS, Ospedale Maggiore, University of
Milan, Milan, Italy
e-mail: [email protected]
Under physiological circumstances, the fat mass, body mass index (BMI) and per-
centage of body fat progressively increase from age 20 and tend to level off at
approximately 80 years in men. By contrast, the fat-free mass increases slightly
from age 20 to 47 and then declines with advancing age [11]. Similar observations
have been reported in other studies, although women have been shown to have
higher percentage body mass values than men across different age groups [12].
Furthermore, the actual age-related increase in fat mass and decrease in lean mass
appear to be somewhat attenuated in women [13]. The possible implications of
gender-associated differences in body composition trajectories across age groups,
both in terms of pharmacokinetics and clinical effects, require further studies.
Left ventricular ejection fraction and cardiac output at rest are both preserved in
healthy older adults. However, the increase in left ventricular ejection fraction that
is normally observed during exercise is blunted in old age, mainly due to a reduction
Table 1.1 Main age-associated structural and functional changes in organs and systems
• Relative increase in body fat and reduction in lean mass
• Blunted cardiac output and heart rate response during exercise
• Reduction in kidney volume and glomerular filtration rate
• Reduction in liver volume and hepatic blood flow
1 The Impact of Ageing on Pharmacokinetics 5
in ejection fraction reserve [14]. Similarly, although the resting heart rate is not
significantly different compared to younger cohorts, a significant reduction in the
maximal, exercise-induced heart rate is observed with advancing age [14]. The lat-
ter is primarily responsible for the age-associated reduction in maximal cardiac out-
put, in the presence of a preserved stroke volume [14]. Therefore, physiological
ageing is not associated with significant changes in cardiac function under resting
conditions. Rather, it is characterised by a diminished overall cardiac reserve and
cardiac output during stress, which might adversely affect the perfusion to critical
organs, for example, the liver and the kidney, involved in drug metabolism and
elimination. The prevalence of heart failure, particularly the subtype with left ven-
tricular dysfunction and reduced ejection fraction, increases significantly with
advancing age [15]. In these patients, the reduced peripheral organ perfusion and
increased venous pressure, with or without concomitant kidney and/or liver disease,
might exert significant effects on pharmacokinetics [16, 17].
1.2.3 Kidney
A progressive volume loss of the kidneys, primarily affecting the cortex, is typically
observed after the age of 50 [18]. There is also a concomitant reduction in glomeru-
lar filtration rate (GFR), approximately 1 mL/min per year, after the age of 40 [19].
These processes can be accelerated by conditions such as hypertension and diabe-
tes, the main causes of chronic kidney disease (CKD) worldwide [20]. Additionally,
age-associated alterations in tubular structure and function can impair the capacity
to reabsorb sodium and secrete hydrogen and potassium, increasing the risk of fluid,
electrolyte and acid–base abnormalities in older adults [21].
There is no convincing evidence that advancing age per se is associated with signifi-
cant changes in gastric emptying, intestinal transit or absorptive capacity of the
small intestine and the colon, despite a reduced gastric secretion of hydrochloride
acid and the presence of morphological changes in enterocytes [22–24]. By con-
trast, a significant age-related reduction, between 20 and 50%, in liver mass and
hepatic blood flow has been reported [25].
1.3.1 Absorption
The absorption process for drugs administered orally predominantly occurs in the
small intestine through passive diffusion or active transport [26]. With advancing
age, drug absorption via passive diffusion across the gastrointestinal epithelium
6 A. A. Mangoni and E. A. Jarmuzewska
1.3.2 Distribution
In line with the previously described age-related changes in body composition, that
is, relative reduction in total body water and lean muscle mass and concomitant
increase in body fat, the volume of distribution of highly lipophilic drugs, for exam-
ple, benzodiazepines, significantly increases in older adults [32]. As a result, there
is a prolongation in the elimination half-life, in absence of significant changes in
clearance (see formula below), with the risk of drug accumulation and toxicity [33]:
0.693 ´ Vd
t½ = ,
CL
where t½ is the half-life, Vd is the volume of distribution and CL is the clearance.
For example, in a study in healthy subjects divided in two age groups (young
adults, n = 7, age 28 ± 4 years; older adults, n = 8, age 69 ± 6 years), the half-life of
diazepam in older adults was nearly three times that of younger participants (86 ± 36
vs. 31 ± 12 h, p < 0.005) [34]. By contrast, the volume of distribution of highly
hydrophilic medications, for example, digoxin, tends to decrease in the older patient
population. In a pharmacokinetic study, the volume of distribution of digoxin in older
and younger participants was 4.1 ± 0.9 L/kg and 5.3 ± 0.6 L/kg, respectively [35].
A progressive reduction in the serum concentrations of the drug-binding protein
albumin has been reported with advancing age [36, 37]. Such changes may theoreti-
cally affect the active, unbound, portion of highly protein-bound drugs, for example,
warfarin and phenytoin. However, the rapid equilibrium occurring through drug elimi-
nation significantly limits this phenomenon’s biological and clinical significance,
therefore, minimising fluctuations in the concentrations of the unbound fraction [38].
1 The Impact of Ageing on Pharmacokinetics 7
1.3.3 Metabolism
The process of drug metabolism occurs primarily in the liver and involves the
biotransformation of active drugs into more soluble inactive or active metabolites
that subsequently undergo renal elimination [39]. These reactions include Phase I
functionalisation (i.e. oxidation, reduction and hydrolysis, mediated by the cyto-
chrome P450 enzyme superfamily) and Phase II conjugation (i.e. acetylation,
glucuronidation and sulphation, mediated by transferases) [40]. Phase I and II
reactions have also been described in the intestine, lung and kidney. However, the
biological and clinical significance of these extra-hepatic reactions are not fully
established [41].
The kidneys are the main site of elimination of soluble drugs and their metabolites.
Renal clearance is considered to be the net result of glomerular filtration, tubular
excretion and tubular reabsorption [57]. The previously reported decline in GFR
with advancing age inevitably leads to impaired drug elimination and consequent
accumulation. This is particularly relevant for drugs that have a narrow therapeutic
index, for example, digoxin, lithium and gentamicin [58]. The dose of such drugs
should be reduced proportionally to the reduction in renal clearance and regularly
monitored [59].
The direct calculation of the GFR by measuring the renal clearance of inulin has
been replaced by formulae that estimate either the creatinine clearance or the GFR
(eGFR), the Cockcroft–Gault formula, the Modification of Diet in Renal Disease
formula and the CKD Epidemiology Collaboration formula [60–62]. However, the
available evidence regarding dose adjustment according to renal function is still
largely based on the Cockcroft–Gault formula.
There is increasing evidence that an impairment in renal function can also influ-
ence absorption, distribution and non-renal elimination. The accumulation of ure-
mic toxins in CKD can inhibit the expression and/or activity of CYP3A4, CYP1A2,
CYP2B6, CYP2C9 and CYP2D6 [63, 64]. Examples of drugs with reduced extra-
renal clearance include carvedilol (CYP3A4, CYP2C9 and CYP2D6), ciprofloxacin
(CYP1A), cyclophosphamide (CYP2B6, CYP2C9 and CYP3A4), duloxetine
(CYP1A and CYP2D6), erythromycin (CYP3A4), solifenacin (CYP3A4) and
tadalafil (CYP3A4) [64]. In some cases, for example, intravenous midazolam
(CYP3A4), the increase in exposure, expressed as area under the concentration
curve, can be as high as sixfold in patients with renal failure when compared with
healthy controls [65].
Table 1.2 Key changes in drug metabolism and elimination in older adults
• Reduction in phase I metabolism, with consequent reduction in first-pass metabolism and
clearance
• Reduction in phase II metabolism, with consequent reduction in clearance, in frail
subjects
• Reduction in the clearance of renally excreted drugs
• Reduction in non-renal drug elimination, through impaired phase I metabolism, in
patients with renal disease
for pre- and/or post-marketing studies that also include frail older participants
(Table 1.3) [70, 69].
1.6 Conclusions
The available evidence suggests that human ageing is characterised by several phar-
macokinetic changes, particularly an increased volume of distribution for highly
lipophilic drugs, a reduction in Phase I metabolism, a reduction in Phase II
10 A. A. Mangoni and E. A. Jarmuzewska
References
1. Qato DM, Wilder J, Schumm LP, Gillet V, Alexander GC. Changes in prescription and
over-the-counter medication and dietary supplement use among older adults in the United
States, 2005 vs 2011. JAMA Intern Med. 2016;176(4):473–82. https://doi.org/10.1001/
jamainternmed.2015.8581.
2. Gao L, Maidment I, Matthews FE, Robinson L, Brayne C, Medical Research Council Cognitive
Function and Ageing Study, et al. Medication usage change in older people (65+) in England
over 20 years: findings from CFAS I and CFAS II. Age Ageing. 2018;47(2):220–5. https://doi.
org/10.1093/ageing/afx158.
3. Bostock CV, Soiza RL, Mangoni AA. Association between prescribing of antimusca-
rinic drugs and antimuscarinic adverse effects in older people. Expert Rev Clin Pharmacol.
2010;3(4):441–52. https://doi.org/10.1586/ecp.10.34.
4. Ruxton K, Woodman RJ, Mangoni AA. Drugs with anticholinergic effects and cognitive
impairment, falls and all-cause mortality in older adults: a systematic review and meta-
analysis. Br J Clin Pharmacol. 2015;80(2):209–20. https://doi.org/10.1111/bcp.12617.
5. Hamzat H, Sun H, Ford JC, Macleod J, Soiza RL, Mangoni AA. Inappropriate prescribing
of proton pump inhibitors in older patients: effects of an educational strategy. Drugs Aging.
2012;29(8):681–90. https://doi.org/10.2165/11632700-000000000-00000.
6. Maes ML, Fixen DR, Linnebur SA. Adverse effects of proton-pump inhibitor use in older
adults: a review of the evidence. Ther Adv Drug Saf. 2017;8(9):273–97. https://doi.
org/10.1177/2042098617715381.
7. Abrahami D, McDonald EG, Schnitzer M, Azoulay L. Trends in prescribing patterns of pro-
ton pump inhibitors surrounding new guidelines. Ann Epidemiol. 2021;55:24–6. https://doi.
org/10.1016/j.annepidem.2020.12.002.
8. Sumukadas D, McMurdo ME, Mangoni AA, Guthrie B. Temporal trends in anticholinergic
medication prescription in older people: repeated cross-sectional analysis of population pre-
scribing data. Age Ageing. 2014;43(4):515–21. https://doi.org/10.1093/ageing/aft199.
9. Reeve E, Wiese MD, Mangoni AA. Alterations in drug disposition in older adults. Expert Opin
Drug Metab Toxicol. 2015;11(4):491–508. https://doi.org/10.1517/17425255.2015.1004310.
10. Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, et al. Frailty in older
adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci. 2001;56(3):M146–56. https://
doi.org/10.1093/gerona/56.3.m146.
11. Jackson AS, Janssen I, Sui X, Church TS, Blair SN. Longitudinal changes in body composi-
tion associated with healthy ageing: men, aged 20–96 years. Br J Nutr. 2012;107(7):1085–91.
https://doi.org/10.1017/S0007114511003886.
12. He X, Li Z, Tang X, Zhang L, Wang L, He Y, et al. Age- and sex-related differences in body
composition in healthy subjects aged 18–82 years. Medicine (Baltimore). 2018;97(25):e11152.
https://doi.org/10.1097/MD.0000000000011152.
1 The Impact of Ageing on Pharmacokinetics 11
13. Bazzocchi A, Diano D, Ponti F, Andreone A, Sassi C, Albisinni U, et al. Health and age-
ing: a cross-sectional study of body composition. Clin Nutr. 2013;32(4):569–78. https://doi.
org/10.1016/j.clnu.2012.10.004.
14. Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular dis-
ease enterprises: part II: the aging heart in health: links to heart disease. Circulation.
2003;107(2):346–54. https://doi.org/10.1161/01.cir.0000048893.62841.f7.
15. Savarese G, Lund LH. Global public health burden of heart failure. Card Fail Rev.
2017;3(1):7–11. https://doi.org/10.15420/cfr.2016:25:2.
16. Mangoni AA, Jarmuzewska EA. The influence of heart failure on the pharmacokinetics of
cardiovascular and non-cardiovascular drugs: a critical appraisal of the evidence. Br J Clin
Pharmacol. 2019;85(1):20–36. https://doi.org/10.1111/bcp.13760.
17. Ergatoudes C, Schaufelberger M, Andersson B, Pivodic A, Dahlstrom U, Fu M. Non-
cardiac comorbidities and mortality in patients with heart failure with reduced vs. pre-
served ejection fraction: a study using the Swedish heart failure registry. Clin Res Cardiol.
2019;108(9):1025–33. https://doi.org/10.1007/s00392-019-01430-0.
18. Dunnill MS, Halley W. Some observations on the quantitative anatomy of the kidney. J Pathol.
1973;110(2):113–21. https://doi.org/10.1002/path.1711100202.
19. Brenner BM, Meyer TW, Hostetter TH. Dietary protein intake and the progressive nature of
kidney disease: the role of hemodynamically mediated glomerular injury in the pathogenesis
of progressive glomerular sclerosis in aging, renal ablation, and intrinsic renal disease. N Engl
J Med. 1982;307(11):652–9. https://doi.org/10.1056/NEJM198209093071104.
20. Sasaki T, Tsuboi N, Okabayashi Y, Haruhara K, Kanzaki G, Koike K, et al. Synergistic impact
of diabetes and hypertension on the progression and distribution of glomerular histopathologi-
cal lesions. Am J Hypertens. 2019;32(9):900–8. https://doi.org/10.1093/ajh/hpz059.
21. Denic A, Glassock RJ, Rule AD. Structural and functional changes with the aging kidney. Adv
Chronic Kidney Dis. 2016;23(1):19–28. https://doi.org/10.1053/j.ackd.2015.08.004.
22. Madsen JL, Graff J. Effects of ageing on gastrointestinal motor function. Age Ageing.
2004;33(2):154–9. https://doi.org/10.1093/ageing/afh040.
23. Dumic I, Nordin T, Jecmenica M, Stojkovic Lalosevic M, Milosavljevic T, Milovanovic
T. Gastrointestinal tract disorders in older age. Can J Gastroenterol Hepatol.
2019;2019:6757524. https://doi.org/10.1155/2019/6757524.
24. Warren PM, Pepperman MA, Montgomery RD. Age changes in small-intestinal mucosa.
Lancet. 1978;2(8094):849–50. https://doi.org/10.1016/s0140-6736(78)92639-9.
25. Wynne HA, Cope LH, Mutch E, Rawlins MD, Woodhouse KW, James OF. The effect of age upon
liver volume and apparent liver blood flow in healthy man. Hepatology. 1989;9(2):297–301.
https://doi.org/10.1002/hep.1840090222.
26. Vertzoni M, Augustijns P, Grimm M, Koziolek M, Lemmens G, Parrott N, et al. Impact of
regional differences along the gastrointestinal tract of healthy adults on oral drug absorp-
tion: an UNGAP review. Eur J Pharm Sci. 2019;134:153–75. https://doi.org/10.1016/j.
ejps.2019.04.013.
27. Maher D, Ailabouni N, Mangoni AA, Wiese MD, Reeve E. Alterations in drug disposi-
tion in older adults: a focus on geriatric syndromes. Expert Opin Drug Metab Toxicol.
2021;17(1):41–52. https://doi.org/10.1080/17425255.2021.1839413.
28. Russell RM. Changes in gastrointestinal function attributed to aging. Am J Clin Nutr. 1992;55(6
Suppl):1203S–7S. https://doi.org/10.1093/ajcn/55.6.1203S.
29. Mangoni AA. The impact of advancing age on P-glycoprotein expression and activity: current
knowledge and future directions. Expert Opin Drug Metab Toxicol. 2007;3(3):315–20. https://
doi.org/10.1517/17425255.3.3.315.
30. Soenen S, Rayner CK, Jones KL, Horowitz M. The ageing gastrointestinal tract. Curr Opin
Clin Nutr Metab Care. 2016;19(1):12–8. https://doi.org/10.1097/MCO.0000000000000238.
31. Jarchow-Macdonald AA, Mangoni AA. Prescribing patterns of proton pump inhibitors in
older hospitalized patients in a Scottish health board. Geriatr Gerontol Int. 2013;13(4):1002–9.
https://doi.org/10.1111/ggi.12047.
12 A. A. Mangoni and E. A. Jarmuzewska
32. Herman RJ, Wilkinson GR. Disposition of diazepam in young and elderly subjects
after acute and chronic dosing. Br J Clin Pharmacol. 1996;42(2):147–55. https://doi.
org/10.1046/j.1365-2125.1996.03642.x.
33. Mangoni AA, Jackson SH. Age-related changes in pharmacokinetics and pharmacodynamics:
basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6–14. https://doi.
org/10.1046/j.1365-2125.2003.02007.x.
34. Greenblatt DJ, Harmatz JS, Zhang Q, Chen Y, Shader RI. Slow accumulation and elimination
of diazepam and its active metabolite with extended treatment in the elderly. J Clin Pharmacol.
2021;61(2):193–203. https://doi.org/10.1002/jcph.1726.
35. Cusack B, Kelly J, O'Malley K, Noel J, Lavan J, Horgan J. Digoxin in the elderly: phar-
macokinetic consequences of old age. Clin Pharmacol Ther. 1979;25(6):772–6. https://doi.
org/10.1002/cpt1979256772.
36. Weaving G, Batstone GF, Jones RG. Age and sex variation in serum albumin concen-
tration: an observational study. Ann Clin Biochem. 2016;53(Pt 1):106–11. https://doi.
org/10.1177/0004563215593561.
37. Gom I, Fukushima H, Shiraki M, Miwa Y, Ando T, Takai K, et al. Relationship between serum
albumin level and aging in community-dwelling self-supported elderly population. J Nutr Sci
Vitaminol (Tokyo). 2007;53(1):37–42. https://doi.org/10.3177/jnsv.53.37.
38. Grandison MK, Boudinot FD. Age-related changes in protein binding of drugs:
implications for therapy. Clin Pharmacokinet. 2000;38(3):271–90. https://doi.
org/10.2165/00003088-200038030-00005.
39. Kinirons MT, O'Mahony MS. Drug metabolism and ageing. Br J Clin Pharmacol.
2004;57(5):540–4. https://doi.org/10.1111/j.1365-2125.2004.02096.x.
40. Klotz U. Pharmacokinetics and drug metabolism in the elderly. Drug Metab Rev.
2009;41(2):67–76. https://doi.org/10.1080/03602530902722679.
41. Gundert-Remy U, Bernauer U, Blomeke B, Doring B, Fabian E, Goebel C, et al. Extrahepatic
metabolism at the body’s internal–external interfaces. Drug Metab Rev. 2014;46(3):291–324.
https://doi.org/10.3109/03602532.2014.900565.
42. Butler JM, Begg EJ. Free drug metabolic clearance in elderly people. Clin Pharmacokinet.
2008;47(5):297–321. https://doi.org/10.2165/00003088-200847050-00002.
43. Pirmohamed M. Prescribing in liver disease. Medicine. 2019;47(11):718–22. https://doi.
org/10.1016/j.mpmed.2019.08.012.
44. Schmucker DL. Liver function and phase I drug metabolism in the elderly: a paradox. Drugs
Aging. 2001;18(11):837–51. https://doi.org/10.2165/00002512-200118110-00005.
45. Le Couteur DG, McLean AJ. The aging liver. Drug clearance and an oxygen dif-
fusion barrier hypothesis. Clin Pharmacokinet. 1998;34(5):359–73. https://doi.
org/10.2165/00003088-199834050-00003.
46. Battino D, Croci D, Mamoli D, Messina S, Perucca E. Influence of aging on serum phenyt-
oin concentrations: a pharmacokinetic analysis based on therapeutic drug monitoring data.
Epilepsy Res. 2004;59(2–3):155–65. https://doi.org/10.1016/j.eplepsyres.2004.04.006.
47. Bebia Z, Buch SC, Wilson JW, Frye RF, Romkes M, Cecchetti A, et al. Bioequivalence revis-
ited: influence of age and sex on CYP enzymes. Clin Pharmacol Ther. 2004;76(6):618–27.
https://doi.org/10.1016/j.clpt.2004.08.021.
48. Herd B, Wynne H, Wright P, James O, Woodhouse K. The effect of age on glucuronidation and
sulphation of paracetamol by human liver fractions. Br J Clin Pharmacol. 1991;32(6):768–70.
49. Court MH. Interindividual variability in hepatic drug glucuronidation: studies into the role of
age, sex, enzyme inducers, and genetic polymorphism using the human liver bank as a model
system. Drug Metab Rev. 2010;42(1):209–24. https://doi.org/10.3109/03602530903209288.
50. Thompson CM, Johns DO, Sonawane B, Barton HA, Hattis D, Tardif R, et al. Database for
physiologically based pharmacokinetic (PBPK) modeling: physiological data for healthy and
health-impaired elderly. J Toxicol Environ Health B Crit Rev. 2009;12(1):1–24. https://doi.
org/10.1080/10937400802545060.
1 The Impact of Ageing on Pharmacokinetics 13
69. Hilmer SN, Kirkpatrick CMJ. New horizons in the impact of frailty on pharmacokinetics: lat-
est developments. Age Ageing. 2021;50(4):1054–63. https://doi.org/10.1093/ageing/afab003.
70. Mangoni AA, Jansen PA, Jackson SH. Under-representation of older adults in pharmaco-
kinetic and pharmacodynamic studies: a solvable problem? Expert Rev Clin Pharmacol.
2013;6(1):35–9. https://doi.org/10.1586/ecp.12.75.
71. Mangoni AA, Pilotto A. New drugs and patient-centred end-points in old age: setting the wheels
in motion. Expert Rev Clin Pharmacol. 2016;9(1):81–9. https://doi.org/10.1586/17512433.201
6.1100074.
Polypharmacy: Definition,
Epidemiology, Consequences 2
and Solutions
She attends her general practitioner (GP) for a routine visit and is noted to be
hypertensive. Amlodipine is prescribed. A few weeks later, she returns to her GP
with bilateral lower limb swelling. The GP prescribes furosemide. She then devel-
ops urinary frequency and is prescribed fesoterodine. She becomes delirious and is
admitted to the hospital. She becomes agitated and is prescribed haloperidol. She
develops constipation, urinary retention and a urinary tract infection. She has sev-
eral non-convulsive vacant episodes which are diagnosed as partial seizures. Her
medications are reviewed, and potentially inappropriate medications are depre-
scribed. Her delirium slowly resolves, and after a short period of rehabilitation, she
is discharged home.
Questions
1. What drug–disease interaction is illustrated in this case?
2. What drug–drug interactions (DDIs) are illustrated in this case?
3. Can you identify the prescribing cascade in this case?
D. Fitzpatrick
Department of Geriatric Medicine, Cork University Hospital, Cork, Ireland
P. F. Gallagher (*)
Department of Geriatric Medicine, Bon Secours Hospital, Cork, Ireland
2.1 Introduction
2.3.1 Pharmacokinetics
2.3.2 Distribution
relative increase in total body fat. This larger Vd results in longer elimination half-
lives and a tendency to accumulation of lipid soluble drugs such as morphine, ben-
zodiazepines, antipsychotics and amitriptyline. This increases the risk of toxicity
and adverse events such as falls and oversedation from these drugs. Smaller doses
and a slower up-titration should therefore be used in older people.
Conversely, the lower lean body mass in older patients means that for water-
soluble drugs, the Vd is smaller than in younger adults leading to higher plasma
concentrations, and therefore, lower doses are necessary. For example, Vd of digoxin
falls with age, and the loading dose should be reduced in older adults [4]. Other
examples of this reduced Vd phenomenon include lithium, theophylline and
gentamicin.
Most drugs are inactive when they are bound to circulating plasma proteins (e.g.
albumin and α-1 glycoprotein). Serum albumin concentration decreases slightly in
older age [20]. More significant reductions in albumin are found in chronic disease
states, which can lead to higher concentrations of unbound (active) drug. This can
result in clinically significant effects for drugs, which are heavily protein bound, for
example, benzodiazepines, antipsychotics, non-steroidal anti-inflammatory drugs
(NSAIDs), warfarin and phenytoin. These drugs have a higher Vd in patients with
hypoalbuminaemia leading to longer half-lives and higher potential for toxicity [19].
2.3.2.1 Metabolism
Liver enzyme activity is preserved in ageing. However, many drug interactions of
patients affected by polypharmacy are mediated through inhibition and induction of
hepatic cytochrome p450 metabolising enzymes. Cytochrome p450 enzyme inhibi-
tion can cause a significant reduction in drug metabolism leading to toxic accumula-
tion. For example, haloperidol inhibits cytochrome p450 2D6, which is responsible
for the metabolism of amitriptyline. Therefore, haloperidol can exacerbate the
potential for anticholinergic side effects of amitriptyline by increasing its active
drug concentration. Similarly, clarithromycin can inhibit the metabolism of statins
leading to statin toxicity, including myositis and hepatitis.
Conversely, drugs such as carbamazepine and phenytoin can induce cytochrome
p450 enzymes, which can accelerate the metabolism and clearance of other drugs.
Clinically relevant examples of cytochrome p450 enzyme inhibitors include antimi-
crobials, antiarrhythmics and some anticoagulants (see Table 2.1) [19].
2.3.2.2 Elimination
The kidneys are primarily responsible for the removal of drugs and their metabolites
from the body. Ageing is associated with reduced renal size, perfusion and urine
concentrating capability, and even in healthy ageing, glomerular filtration rate
(GFR) begins to decline progressively from the age of 30 years onwards [21].
Chronic diseases such as hypertension and diabetes mellitus and the use of nephro-
toxic medications such as NSAIDs accelerate this decline in GFR.
Serum creatinine concentration should not be used as the sole measure to esti-
mate renal function in older adults, because production of creatinine is related to
muscle mass which, as discussed earlier, is significantly reduced in older adults.
Approximately 50% of older patients have a normal serum creatinine level but a
reduced creatinine clearance estimate. The Cockcroft–Gault formula [22], based on
serum creatinine, age, weight and sex, has been the most common method of esti-
mating GFR. This formula underestimates GFR in older adults and newer formulas
such as the CKD Epidemiology Collaboration (CKD-EPI, based on serum creati-
nine, age and sex) [23], are likely more accurate in older individuals. However,
many dosing recommendations are based on the Cockcroft–Gault formula.
Older adults frequently develop acute kidney injury (AKI) in the context of acute
illness and premorbid chronic kidney disease (CKD). Precipitating factors include
hypotension, dehydration, sepsis, cardiorenal syndrome and cardiac failure. Drugs
often mediate or exacerbate AKI in older adults especially NSAIDs, diuretics, ACE
inhibitors and angiotensin receptor blockers. These drugs often need to be stopped
temporarily in patients with an AKI or those at high risk of an AKI. Other drugs may
need to be reduced or stopped because of the risk of toxic accumulation during an
AKI or in the context of CKD, for example, lithium and metformin.
2.3.3 Pharmacodynamics
Polypharmacy
Hospitalisation
Reduced Quality of Life
Mortality
18 billion US dollars, 0.3% of the global total health expenditure, could be saved by
avoidance of inappropriate prescribing [37]. Avoiding ADEs would not only
improve safety outcomes for patients but would also reduce costs in terms of fewer
hospital admissions, readmissions and shorter length of stay. National campaigns
promoting appropriate prescribing and reducing inappropriate polypharmacy have
demonstrated significant cost savings and economic benefits in Sweden [38] and
Scotland [39] (Fig. 2.1).
reconciliation. This is all the more likely in older patients who may be on many dif-
ferent medications and are not able to reliably discuss their medication regimens.
Transitions in care represent an opportunity for medication errors with potentially
serious adverse outcomes to occur [41, 42]. Careful medication reconciliation is
vital to reduce these errors. Medication reconciliation is dealt with in more detail in
Chap. 8.
2.3.5.3 Adherence
Non-adherence with prescribed medication is a major public health issue and is
closely associated with polypharmacy [17]. It is estimated that between 50 and 80%
of patients with chronic conditions are non-adherent with their medications. Non-
adherence has been estimated to contribute to 48% of asthma deaths, an 80%
increased risk of death in diabetes and a 3.8-fold increased mortality following
myocardial infarction [43]. It is estimated that non-adherence to medicines costs the
European Union 125 billion euros per year [17].
Adherence may be intentional or nonintentional. Barriers to non-adherence
should be addressed. For example, a patient with cognitive impairment may simply
forget to take their medications or make mistakes in their administration. Medication
blister packs or reminder alarms are simple ways to overcome some to these barriers
(Fig. 2.2).
Frailty and polypharmacy are clearly linked in a bidirectional way, that is, frailty
reflects multimorbidity, which generates polypharmacy, whilst polypharmacy
engenders PIMs and ADRs/ADEs which often exacerbate frailty. The severely frail
older person cohort is especially vulnerable to the adverse effects of polypharmacy
[47]. The goals of medication in this group are often directed toward symptomatic
benefit rather than prolonging life and long-term disease prevention. Despite this,
the SHELTER study reported rates of polypharmacy (defined as 5–9 long-term
drugs daily) and excessive polypharmacy (≥10 drugs) in nursing home residents to
be 48.7% and 24.3%, respectively [44].
In patients with frailty and limited life expectancy, medication review should
focus on reducing medication burden and optimising symptom management, rather
than long-term preventative strategies. The STOPPFrail criteria provide a list of
medications that should be discontinued or withdrawn in certain settings for older
people who meet all the following criteria: ‘end stage irreversible pathology, poor
1-year survival prognosis, severe functional impairment or severe cognitive impair-
ment or both, [in whom] symptom control is the priority rather than prevention of
disease progression’ [48, 49].
Whilst medication review tools are helpful in this group, it is important to recog-
nise that this is a heterogeneous group and prescribing decisions should be individu-
alised, and the patient (and carer when appropriate) should be involved as much as
possible.
Author Declaration The above chapter is, in its entirety, the work of the listed authors. It does
not contain any third party material or any material under copyright.
References
1. Corselli-Nordblad L, Strandell H. Ageing Europe—Looking at the lives of older people in the
EU 2020 edition. Luxembourg: Eurostat; 2020. https://doi.org/10.2785/628105.
2. United Nations Department of Economic and Social Affairs Population Division. The world
population ageing 2020 highlights: living arrangements of older persons. New York: United
Nations; 2020.
3. Wastesson JW, Morin L, Tan ECK, Johnell K. An update on the clinical consequences of poly-
pharmacy in older adults: a narrative review. Expert Opin Drug Saf. 2018;17:1185–96. https://
doi.org/10.1080/14740338.2018.1546841.
4. Reeve E, Wiese MD, Mangoni AA. Alterations in drug disposition in older adults. Expert Opin
Drug Metab Toxicol. 2015;11:491–508.
5. Maher D, Ailabouni N, Mangoni AA, Wiese MD, Reeve E. Alterations in drug disposition in
older adults: a focus on geriatric syndromes. Expert Opin Drug Metab Toxicol. 2020;17:41–52.
https://doi.org/10.1080/17425255.2021.1839413.
6. Ibanez B, James S, Agewall S, et al. 2017 ESC guidelines for the management of acute myo-
cardial infarction in patients presenting with ST-segment elevation. The task force for the man-
agement of acute myocardial infarction in patients presenting with ST-segment elevation of the
European Society of Cardiology (ESC). Eur Heart J. 2018;39:119–77.
7. Scottish Government Polypharmacy Model of Care Group. Polypharmacy guidance, realistic
prescribing. 3rd ed. Edinburgh: NHS; 2018.
8. Kantor ED, Rehm CD, Haas JS, Chan AT, Giovannucci EL. Trends in prescription drug use
among adults in the United States from 1999 to 2012. JAMA. 2015;314:1818–30.
9. Khezrian M, McNeil CJ, Murray AD, Myint PK. An overview of prevalence, determinants and
health outcomes of polypharmacy. Ther Adv Drug Saf. 2020;11:2042098620933741. https://
doi.org/10.1177/2042098620933741.
10. Moriarty F, Hardy C, Bennett K, Smith SM, Fahey T. Trends and interaction of polypharmacy
and potentially inappropriate prescribing in primary care over 15 years in Ireland: a repeated
cross-sectional study. BMJ Open. 2015;5:e008656.
11. Midão L, Giardini A, Menditto E, et al. Polypharmacy prevalence among older adults based on
the survey of health, ageing and retirement in Europe. Arch Gerontol Geriatr. 2018;78:213–20.
12. Wastesson JW, Fastbom J, Johnell K. Expanding the proportion of life with polypharmacy in
Sweden: 2006–2013. J Am Med Dir Assoc. 2016;17:957–8.
13. Hernández-Rodríguez MÁ, Sempere-Verdú E, Vicens-Caldentey C, et al. Evolution of poly-
pharmacy in a Spanish population (2005–2015): a database study. Pharmacoepidemiol Drug
Saf. 2020;29:433–43.
30 D. Fitzpatrick and P. F. Gallagher
14. Lukačišinová A, Fialová D, Peel NM, et al. The prevalence and prescribing patterns of benzo-
diazepines and Z-drugs in older nursing home residents in different European countries and
Israel: retrospective results from the EU SHELTER study. BMC Geriatr. 2021;211(21):1–16.
15. Toepfer S, Bolbrinker J, König M, Steinhagen-Thiessen E, Kreutz R, Demuth I. Potentially
inappropriate medication in older participants of the Berlin Aging Study II (BASE-II)—sex dif-
ferences and associations with morbidity and medication use. PLoS One. 2019;14:e0226511.
16. Franchi C, Tettamanti M, Pasina L, Djignefa CD, Fortino I, Bortolotti A, Merlino L, Nobili
A. Changes in drug prescribing to Italian community-dwelling elderly people: the EPIFARM-
elderly project 2000–2010. Eur J Clin Pharmacol. 2014;70:437–43.
17. Mair A, Fernandez-Llimos F, Alonso A, Harrison C, Hurding S, Kempen T, Kinnear M,
Michael N, McIntosh J, Wilson M. Polypharmacy management by 2030: a patient safety chal-
lenge. 2nd ed. Coimbra: Simpathy; 2017.
18. Kongkaew C, Hann M, Mandal J, Williams SD, Metcalfe D, Noyce PR, Ashcroft DM. Risk
factors for hospital admissions associated with adverse drug events. Pharmacotherapy.
2013;33:827–37.
19. Mangoni AA, Jackson SHD. Age-related changes in pharmacokinetics and pharmacodynam-
ics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57:6.
20. Weaving G, Batstone GF, Jones RG. Age and sex variation in serum albumin con-
centration: an observational study. Ann Clin Biochem. 2015;53:106–11. https://doi.
org/10.1177/0004563215593561.
21. Glassock RJ, Winearls C. Ageing and the glomerular filtration rate: truths and consequences.
Trans Am Clin Climatol Assoc. 2009;120:419.
22. Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron.
1976;16:31–41.
23. Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration
rate. Ann Intern Med. 2009;150:604–12.
24. Johnell K, Klarin I. The relationship between number of drugs and potential drug–drug inter-
actions in the elderly: a study of over 600,000 elderly patients from the Swedish Prescribed
Drug Register. Drug Saf. 2007;30:911–8.
25. de Oliveira LM, Diel JD, Nunes A, Dal Pizzol TD. Prevalence of drug interactions in hospital-
ised elderly patients: a systematic review. Eur J Hosp Pharm. 2021;28:4–9.
26. Danijea G, Kristina J. Clinical implications from drug–drug and drug–disease interactions in
older people. Clin Exp Pharmacol Physiol. 2013;40:320–5.
27. Fried TR, O’Leary J, Towle V, Goldstein MK, Trentalange M, Martin DK. Health outcomes
associated with polypharmacy in community-dwelling older adults: a systematic review. J Am
Geriatr Soc. 2014;62:2261–72.
28. Laflamme L, Monárrez-Espino J, Johnell K, Elling B, Möller J. Type, number or both? A
population-based matched case-control study on the risk of fall injuries among older people
and number of medications beyond fall-inducing drugs. PLoS One. 2015;10(3):e0123390.
https://doi.org/10.1371/JOURNAL.PONE.0123390.
29. Ambrose AF, Paul G, Hausdorff JM. Risk factors for falls among older adults: a review of the
literature. Maturitas. 2013;75:51–61.
30. Compston J, Cooper A, Cooper C, et al. UK clinical guideline for the prevention and treatment
of osteoporosis. Arch Osteoporos. 2017;12:43.
31. Niikawa H, Okamura T, Ito K, Ura C, Miyamae F, Sakuma N, Ijuin M, Inagaki H, Sugiyama
M, Awata S. Association between polypharmacy and cognitive impairment in an elderly
Japanese population residing in an urban community. Geriatr Gerontol Int. 2017;17:1286–93.
32. Rawle MJ, Cooper R, Kuh D, Richards M. Associations between polypharmacy and cognitive
and physical capability: a British birth cohort study. J Am Geriatr Soc. 2018;66:916–23.
33. Fox C, Richardson K, Maidment ID, Savva GM, Matthews FE, Smithard D, Coulton S, Katona
C, Boustani MA, Brayne C. Anticholinergic medication use and cognitive impairment in the
older population: the Medical Research Council Cognitive Function and Ageing Study. J Am
Geriatr Soc. 2011;59:1477–83.
2 Polypharmacy: Definition, Epidemiology, Consequences and Solutions 31
34. Welsh TJ, van der Wardt V, Ojo G, Gordon AL, Gladman JRF. Anticholinergic drug burden
tools/scales and adverse outcomes in different clinical settings: a systematic review of reviews.
Drugs Aging. 2018;356(35):523–38.
35. Drake MJ, Nixon PM, Crew JP. Drug-induced bladder and urinary disorders. Drug Saf.
1998;19:45–55.
36. Calderón-Larrañaga A, Poblador-Plou B, González-Rubio F, Gimeno-Feliu LA, Abad-Díez
JM, Prados-Torres A. Multimorbidity, polypharmacy, referrals, and adverse drug events: are
we doing things well? Br J Gen Pract. 2012;62:e821–6.
37. Aitken M, Gorokhovich L. Advancing the responsible use of medicines: applying levers for
change. SSRN Electron J. 2012;2012:2222541. https://doi.org/10.2139/SSRN.2222541.
38. Kempen TGH, Bertilsson M, Hadziosmanovic N, Lindner K-J, Melhus H, Nielsen EI, Sulku
J, Gillespie U. Effects of hospital-based comprehensive medication reviews including postdis-
charge follow-up on older patients’ use of health care: a cluster randomized clinical trial. JAMA
Netw Open. 2021;4(4):e216303. https://doi.org/10.1001/JAMANETWORKOPEN.2021.6303.
39. Mair A, Wilson M, Dreischulte T. The polypharmacy programme in Scotland: realistic pre-
scribing. Prescriber. 2019;30:10–6.
40. Rochon PA, Petrovic M, Cherubini A, Onder G, O’Mahony D, Sternberg SA, Stall NM,
Gurwitz JH. Polypharmacy, inappropriate prescribing, and deprescribing in older people:
through a sex and gender lens. Lancet Health Longev. 2021;2:e290–300.
41. Wheeler AJ, Scahill S, Hopcroft D, Stapleton H. Reducing medication errors at transitions of
care is everyone’s business. Aust Prescr. 2018;41:73.
42. Gleason KM, McDaniel MR, Feinglass J, Baker DW, Lindquist L, Liss D, Noskin GA. Results
of the medications at transitions and clinical handoffs (MATCH) study: an analysis of
medication reconciliation errors and risk factors at hospital admission. J Gen Intern Med.
2010;25:441–7.
43. Rachel E. Non-adherence to medicines: not solved but solvable. J Health Serv Res Policy.
2009;14:58–61.
44. Paul G, Cristin R, Stephen B, Julia K, Denis O. STOPP (screening tool of older person’s pre-
scriptions) and START (screening tool to alert doctors to right treatment). Consensus valida-
tion. Int J Clin Pharmacol Ther. 2008;46:72–83.
45. O’Mahony D, O’Sullivan D, Byrne S, O’Connor MN, Ryan C, Gallagher P. STOPP/START
criteria for potentially inappropriate prescribing in older people: version 2. Age Ageing.
2015;44:213–8.
46. O’Connor MN, O’Sullivan D, Gallagher PF, Eustace J, Byrne S, O’Mahony D. Prevention of
hospital-acquired adverse drug reactions in older people using screening tool of older persons’
prescriptions and screening tool to alert to right treatment criteria: a cluster randomized con-
trolled trial. J Am Geriatr Soc. 2016;64:1558–66.
47. Lavan AH, Gallagher P. Predicting risk of adverse drug reactions in older adults. Ther Adv
Drug Saf. 2016;7:11.
48. Lavan AH, Gallagher P, Parsons C, O’Mahony D. STOPPFrail (screening tool of older persons
prescriptions in frail adults with limited life expectancy): consensus validation. Age Ageing.
2017;46:600–7.
49. Curtin D, Gallagher P, O’Mahony D. Deprescribing in older people approaching end-of-life:
development and validation of STOPPFrail version 2. Age Ageing. 2021;50:465–71.
Drug–Drug and Drug–Nutrients
Interactions: From Theory to Clinical 3
Relevance
3.1 Introduction
E. M. de Koning
Department of Hospital Pharmacy, Medisch Spectrum Twente, Enschede, The Netherlands
e-mail: [email protected]
J. Huisbrink
Department of Hospital Pharmacy, Franciscus Gasthuis en Vlietland,
Schiedam/Rotterdam, The Netherlands
e-mail: [email protected]
W. Knol (*)
Department of Geriatric Medicine and Expertise Centre Pharmacotherapy in Old Persons,
University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
e-mail: [email protected]
Not only interactions between drugs are prevalent. Drugs can also interfere with
certain foods or enteral feeding (drug–nutrition interactions or DNIs), herbs or flu-
ids like alcohol. Drug–nutrition interactions are defined as modifications of pharma-
cokinetics or pharmacodynamics of a drug because of nutrients or a modification in
nutritional status because of the addition of a drug [12].
This chapter will focus on drug–drug interactions and drug–nutrition interactions.
3.2 Theory
3.2.1.1 Absorption
Absorption defines the uptake of a substance into the systemic circulation. Several
factors influence the degree of absorption, such as physiochemical properties (e.g.
drug solubility, lipophilicity of the drug), drug formulation, route of administration
and patient factors (e.g. gastrointestinal pH). Additionally, the presence of food
could affect drug absorption, for example, by decreasing gastric emptying, chang-
ing the pH of the stomach and decreasing gut motility [13–15].
An active substance can become unsuitable for absorption due to chemical reac-
tion with another active substance within the gastrointestinal tract. This happens for
example when iron is given simultaneously with quinolones, such as ciprofloxacin.
The latter drugs form an insoluble complex, which reduces the bioavailability of
ciprofloxacin. This interaction could be avoided by prolonging the interval of
3 Drug–Drug and Drug–Nutrients Interactions: From Theory to Clinical Relevance 35
3.2.1.2 Distribution
The absorbed drug will be distributed after entering the general circulation. Drugs
can bind to several proteins in the bloodstream or diffuse to tissues located outside
the bloodstream. Many drugs are highly bound to plasma proteins, such as albumin
or alpha-1-acid glycoprotein. An interaction may emerge if one agent displaces
another agent from its plasma binding site. By displacement, more unbound drug
becomes available. Such protein binding interactions hardly ever lead to clinically
significant changes in efficacy, since there will be further distribution or elimination
of the drug. In theory, the interaction could become relevant if one agent has a small
volume of distribution, a narrow therapeutic index and high plasma protein binding
properties.
3.2.1.3 Metabolism
The cytochrome P450 (CYP) enzyme family plays a very important role in catalys-
ing the biotransformation of certain drugs and other xenobiotic agents. Approximately
half of the 200 most commonly used drugs undergo CYP-mediated metabolism
[17]. CYP3A4/5, CYP2D6, CYP2C9, CYP2B6 and CYP2C19 are mostly involved
in this metabolism [18].
Drug metabolism occurs mainly within the liver and kidney, but could also take
place inside the lungs or gastro-intestinal tract, where CYP3A4 is present in the
gut wall.
36 E. M. de Koning et al.
3.2.1.4 Elimination
Elimination is a process of clearance of the drug, mostly by the kidney or via bile.
There are also other routes of elimination, such as pulmonary or dermal
elimination.
Drug-induced changes in hepatic blood flow could affect certain drugs that have
high hepatic extraction ratios, such as propranolol. Drugs that increase hepatic
blood flow, such as glucagon and verapamil, accelerate elimination of drugs like
propranolol. This, however, has an unknown clinical relevance.
Renal elimination of drugs may be diminished by agents that decrease glomerular
filtration rate, such as aminoglycoside antibiotics or angiotensin-converting enzyme
(ACE)-inhibitors. Drug-induced renal impairment may be aggravated by concomi-
tantly prescribing drugs that enhance renal impairment. A combination of non-steroi-
dal anti-inflammatory drugs (NSAIDs) and ACE-inhibitors could lead to renal failure.
A third mechanism that could lead to interactions is competition for tubular
active transport. One substance could disrupt renal excretion of another agent, pos-
sibly leading to accumulation and toxicity. For example, the body recognizes lith-
ium as sodium and is, therefore, processed in similar ways. Drugs that induce
reabsorption of sodium could also provoke reabsorption of lithium, resulting in
toxicity.
3 Drug–Drug and Drug–Nutrients Interactions: From Theory to Clinical Relevance 37
There are minor changes regarding absorption in the older patient. The absorption
is slightly slowed, but total drug absorption does not change. There is a higher avail-
ability for drugs with very large first pass effect. The gastric pH is higher, which
could lead to a variable absorption of tablets requiring low pH. However, most of
these changes do not lead to clinically relevant alterations in drug levels.
Older patients hold more fat, less muscle tissue and less body water. Hydrophilic
drugs with a high volume of distribution (e.g. digoxin) have a smaller volume of
distribution due to less fluid and muscle tissue. Thus, a lower loading dose of
38 E. M. de Koning et al.
digoxin should be given to avoid toxicity. On the contrary, lipophilic drugs have a
larger volume of distribution. Therefore, accumulation of lipophilic drugs (e.g.
diazepam) could occur, and such drugs may cause prolongation of action and
adverse effects.
Phase I metabolism is reduced in older people (approximately 30%), especially
regarding CYP1A2 and CYP3A4. Phase II metabolism remains intact. Clinical con-
sequences regarding altered phase I metabolism remain unknown.
The age-related changes concerning elimination are of highest relevance. Kidney
function decreases with age but is also affected by comorbidities such as hyperten-
sion or diabetes. In case of renal insufficiency, dose adjustments are frequently nec-
essary for drugs that are cleared renally.
PK interactions that could become clinically relevant are mostly due to CYP-related
metabolism. For example, the use of ciprofloxacin (CYP1A2 inhibitor) in older
patients treated with clozapine (CYP1A2 substrate) leads to a decreased metabo-
lism of clozapine. Moreover, phase I metabolism by CYP1A2 is shown to be
reduced by aging. Additionally, CYP1A2 activity is known to be reduced during
infection. These combined effects may further enhance the therapeutic effects or
even toxicity of clozapine.
Another example is the combination of simvastatin or atorvastatin and CYP3A4
inhibitors. Increased statin exposure due to the inhibition of CYP3A4-mediated
metabolism by, for example, verapamil and grapefruit juice could lead to severe
myopathy and rhabdomyolysis, which, in turn, may lead to acute renal failure. On
the contrary, prescribing simvastatin with CYP3A4 inducers such as carbamazepine
(70–80% reduction of plasma concentration) or St John’s wort (60% reduction)
could lead to higher cholesterol concentrations [19, 20].
A third example is the combination of lithium with an ACE-inhibitor or
diuretic. Diuretics or ACE-inhibitors enhance sodium loss. Because of this, there
will be a compensatory distal reabsorption of sodium and lithium, leading to lith-
ium toxicity.
Type of
Drug class Drug/nutrient Drug/nutrient Impact/mechanism IA
Cardiovascular ACE-inhibitors Potassium supplements/ Elevated serum potassium PD
aldosterone antagonists/
potassium sparing diuretics
ACE-inhibitor ARB Risk of renal failure by additive or synergistic effects on PD
renal blood flow and blood pressure
Digoxin Amiodarone Digoxin toxicity due to p-gp inhibition; both drugs PK + PD
suppress the SA node, resulting in bradycardia
Digoxin Verapamil PK: Verapamil inhibits both renal tubular secretion and PK + PD
non-renal excretion of digoxin. Diltiazem decreases the
clearance and/or volume of distribution of digoxin
PD: The slowing effects of verapamil or diltiazem and
digoxin on atrioventricular conduction may be additive
Verapamil/diltiazem CYP3A4 substrates (e.g. Inhibition metabolism of substrates, therefore higher PK
carbamazepine, simvastatin, concentrations
midazolam)
Dabigatran/edoxaban P-gp inhibitor Higher risk of bleeding PK
Rivaroxaban/apixaban P-gp inhibitor or CYP3A4 Higher risk of bleeding PK
inhibitor
Oral anticoagulant Antiplatelet drug Higher risk of bleeding PD
Antiplatelet drugs Ginkgo biloba Increased risk of bleeding. Mechanism remains unknown PD
Beta-blocker Verapamil/diltiazem Increased effect on AV nodal conduction, heart rate or PD
cardiac contractility, resulting in bradycardia, AV block
and hypotension
Beta-blocker Beta2-agonist Beta-blockers may diminish the bronchodilatory effect of PD
beta2-agonists. Of particular concern with non-selective
beta-blockers or higher doses of the beta1 selective
beta-blockers
E. M. de Koning et al.
Type of
Drug class Drug/nutrient Drug/nutrient Impact/mechanism IA
3
(continued)
41
Table 3.1 (continued)
42
Type of
Drug class Drug/nutrient Drug/nutrient Impact/mechanism IA
MAO-inhibitors SSRI/opioid Serotonin syndrome or opioid toxicity by enhancing the PD
serotonergic effect of MAO inhibitors
MAO-inhibitors Tyramine-rich foods Severe hypertensive episodes by inhibiting tyramine PK
metabolism, leading to increased biosynthesis of
catecholamines
Phenytoin Enteral nutrition formulations Phenytoin binds proteins and calcium salts in enteral PK
nutrition formulations, leading to reduced absorption
Paroxetine/fluoxetine Metoprolol Increase in metoprolol concentrations due to CYP2D6 PK
inhibition, possibly resulting in bradycardia
Fluoxetine Tricyclic antidepressant Risk of serotonin syndrome by additive serotonergic PK + PD
effects or tricyclic antidepressant toxicity due to
fluoxetine-mediated inhibition of CYP2D6 and/or
CYP2C19, enzymes responsible for the metabolism of
tricyclic antidepressants
Paroxetine/fluoxetine St John’s wort St John’s wort may enhance the serotonergic effect of PD
paroxetine or fluoxetine, which could result in serotonin
syndrome
Concomitant use of ≥3 centrally acting drugs (e.g. opioids, Increased risk of falling, possibly leading to fractures and PD
antipsychotics, benzodiazepines, antidepressants or impaired cognition
antiepileptics)
Clozapine CYP1A2 inducers/inhibitors Influences on clozapine concentrations due to induction or PK
(e.g. carbamazepine/ inhibition of CYP1A2, leading to loss of effect or toxicity
ciprofloxacin)
Anti-infective agents Fluoroquinolones/doxycyclin Calcium/dairy products/iron/ Reduced absorption due to complexation PK
cations in enteral nutrition
Antifungal agents e.g. Warfarin/acenocoumarol/ Increased serum concentrations of vitamin K antagonists PK
miconazole fenprocoumon by inhibition of CYP2C9 enzymes
E. M. de Koning et al.
Type of
Drug class Drug/nutrient Drug/nutrient Impact/mechanism IA
3
Type of
Drug class Drug/nutrient Drug/nutrient Impact/mechanism IA
Gastro-intestinal Proton pump inhibitor Vitamin B12 Malabsorption of vitamin B12 due to inhibition of PK
intrinsic factor
Sucralfate Enteral nutrition formulations Sucralfate binds proteins within the enteral feed, leading PC
to a change in consistency and possible feeding tube
blockage
Other Bisphosphonate Calcium/dairy products/iron/ Reduced absorption due to complexation PK
cations in enteral nutrition
St John’s wort CYP3A4 substrate (e.g. Reduced efficacy due to induction of CYP3A4 PK
verapamil)
ACE angiotensin-converting enzyme, ARB angiotensin II receptor blocker, AV atrioventricular, AUC area under the curve, BBB blood–brain barrier, CYP cyto-
chrome P450, D2 dopamine 2 receptor, IA interaction, MAO monoamine oxidase, NSAID non-steroidal anti-inflammatory drug, PC pharmaceutical, PD phar-
macodynamic, P-gp P-glycoprotein, PK pharmacokinetic, RAAS renin–angiotensin–aldosterone-system, SA sinoatrial, SNRI serotonin and noradrenalin
reuptake inhibitor, SSRI selective serotonin reuptake inhibitor
E. M. de Koning et al.
3 Drug–Drug and Drug–Nutrients Interactions: From Theory to Clinical Relevance 45
References
1. Masnoon N, Shakib S, Kalisch-Ellett L, Caughey GE. What is polypharmacy? A sys-
tematic review of definitions. BMC Geriatr. 2017;17(1):230. https://doi.org/10.1186/
s12877-017-0621-2.
2. Salazar JA, Poon I, Nair M. Clinical consequences of polypharmacy in elderly: expect the
unexpected, think the unthinkable. Expert Opin Drug Saf. 2007;6(6):695–704.
3. Shetty V, Chowta MN, Chowta KN, Shenoy A, Kamath A, Kamath P. Evaluation of potential
drug–drug interactions with medications prescribed to geriatric patients in a tertiary care hos-
pital. J Aging Res. 2018;2018:5728957.
4. Bjerrum L, Andersen M, Petersen G, Kragstrup J. Exposure to potential drug interactions in
primary health care. Scand J Prim Health Care. 2003;21:153–8.
5. Magro L, Moretti U, Leone R. Epidemiology and characteristics of adverse drug reactions
caused by drug–drug interactions. Expert Opin Drug Saf. 2012;11:83–94.
6. Mallet L, Spinewine A, Huang A. The challenge of managing drug interactions in elderly
people. Lancet. 2007;370:185–91.
7. Hines LE, Murphy JE. Potentially harmful drug–drug interactions in the elderly: a review. Am
J Geriatr Pharmacother. 2011;9:364–77.
8. Gnjidic D, Johnell K. Clinical implications from drug–drug and drug–disease interactions in
older people. Clin Exp Pharmacol Physiol. 2013;40:320–5.
9. Bykov K, Gagne JJ. Generating evidence of clinical outcomes of drug–drug interactions. Drug
Saf. 2017;40:101–3.
10. Swart F, Bianchi G, Lenzi J, Iommi M, Maestri L, Raschi E, et al. Risk of hospitalization from
drug–drug interactions in the elderly: real-world evidence in a large administrative database.
Aging. 2020;12(19):711–39.
11. Dechanont S, Maphanta S, Butthum B, Kongkaew C. Hospital admissions/visits associated
with drug–drug interactions: a systematic review and meta-analysis. Pharmacoepidemiol Drug
Saf. 2014;23:489–97.
12. Genser D. Food and drug interaction: consequences for the nutrition/health status. Ann Nutr
Metab. 2008;52:29–32.
13. Amadi CN, Mgbahurike AA. Selected food/herb–drug interactions: mechanisms and clinical
relevance. Am J Ther. 2018;25(4):e423–33.
14. Akamine D, Filho MK, Peres CM. Drug–nutrient interactions in elderly people. Curr Opin
Clin Nutr Metab Care. 2007;10(3):304–10.
15. Briguglio M, Hrelia S, Malaguti M, Serpe L, Canaparo R, Dell'Osso B, Galentino R, De
Michele S, Dina CZ, Porta M, Banfi G. Food bioactive compounds and their interference in
drug pharmacokinetic/pharmacodynamic profiles. Pharmaceutics. 2018;10(4):277.
16. White R, Bradnam V. Handbook of drug administration via enteral feeding tubes. London:
Pharmaceutical Press; 2015.
17. Williams JA, Hyland R, Jones BC, Smith DA, Hurst S, Goosen TC, et al. Drug–drug interac-
tions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically
observed low exposure (AUCi/AUC) ratios. Drug Metab Dispos. 2004;32(11):1201–8.
18. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of
gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther.
2013;138(1):103–41.
19. Ucar M, et al. Carbamazepine markedly reduces serum concentrations of simvastatin and sim-
vastatin acid. Eur J Clin Pharmacol. 2004;59:879–82.
20. Eggertsen R, et al. Effects of treatment with a commercially available St John’s Wort product
(Movina) on cholesterol levels in patients with hypercholesterolemia treated with simvastatin.
Scand J Prim Health Care. 2007;25:154–9.
21. Bowie MW, Slattum PW. Pharmacodynamics in older adults: a review. Am J Geriatr
Pharmacother. 2007;5(3):263–303.
46 E. M. de Koning et al.
22. Vestal RE, Wood AJJ, Shand DG. Reduced β-adrenoceptor sensitivity in the elderly. Clin
Pharmacol Ther. 1979;26(2):181–6.
23. Ferrara N, Komici K, Corbi G, Pagano G, Furgi G, Rengo C, Femminella GD, Leosco D,
Bonaduce D. β-Adrenergic receptor responsiveness in aging heart and clinical implications.
Front Physiol. 2014;9(4):396.
24. White M, Roden R, Minobe W, Khan MF, Larrabee P, Wollmering M, et al. Age-related changes
in β-adrenergic neuroeffectors system in the human heart. Circulation. 1994;90:1225–38.
25. Scarpace PJ, Turner N, Mader SL. β-Adrenergic function in aging. Drugs Aging. 1991;1:116–29.
26. Zerah L, Henrard S, Wilting I, O'Mahony D, Rodondi N, Dalleur O, Dalton K, Knol W,
Haschke M, Spinewine A. Prevalence of drug–drug interactions in older people before and
after hospital admission: analysis from the OPERAM trial. BMC Geriatr. 2021;21(1):571.
27. Tulner LR, Frankfort SV, Gijsen GJ, van Campen JP, Koks CH, Beijnen JH. Drug–drug
interactions in a geriatric outpatient cohort: prevalence and relevance. Drugs Aging.
2008;25(4):343–55.
28. Anrys P, Petit AE, Thevelin S, Sallevelt B, Drenth C, Soiza RL, Correa-Pérez A, Dalleur O, De
Brauwer I, Petrovic M, Coleman JJ, Dalton K, O'Mahony D, Löwe A, Thürig S, Gudmundsson
A, Cherubini A, Panos A, Mavridis D, Rodondi N, Spinewine A. An international consensus
list of potentially clinically significant drug–drug interactions in older people. J Am Med Dir
Assoc. 2021;22(10):2121–2133.e24.
29. Vonbach P, Dubied A, Krähenbühl S, Beer JH. Prevalence of drug–drug interactions at
hospital entry and during hospital stay of patients in internal medicine. Eur J Intern Med.
2008;19(6):4123–0.
Inappropriate Prescription of Medicines
4
Denis O’Mahony
4.1 Introduction
Inappropriate prescribing (IP) of medication to patients of any age refers to any one
or more of the following patterns:
Instances of IP that fall into categories (1)–(5) are generally referred to as ‘poten-
tially inappropriate medications’ or PIMs; instances of category (6) are termed
‘potential prescribing omissions’ or PPOs.
PIMs and PPOs are important in older people because they are associated with
significant adversity, that is, increased risk of adverse drug reactions (ADRs) and
adverse drug events (ADEs), greater levels of unscheduled hospitalization, more
healthcare utilization (mostly through primary care physician consultation), poorer
quality of life and increased mortality. It is estimated that more than half of all
instances of PIMs and PPOs are avoidable; this has highly significant economic, as
D. O’Mahony (*)
Department of Medicine, University College Cork, Cork, Ireland
Department of Geriatric Medicine, Cork University Hospital, Cork, Ireland
e-mail: [email protected]
There are several sets of criteria for defining IP in older people in the literature.
These are broadly classified as ‘implicit criteria’ and ‘explicit criteria’.
Implicit IP criteria are usually based on decision algorithms without specific
reference to any one drug or drug class; well-cited examples include ACOVE crite-
ria and the Medication Appropriateness Index (MAI) [1, 2]. In general, implicit IP
criteria have remained in the research domain and have not been deployed in routine
clinical practice to any significant extent.
Explicit IP criteria, in contrast, have had much greater application in the clinical
arena. The first of the explicit IP criteria sets was published in 1991 by Mark Beers
and colleagues [3]. This first iteration of Beers criteria provided for the first time a
list of drugs and drug doses that should be avoided in older nursing home residents
in all circumstances and certain drugs that should be avoided in certain disease
groups. Since the first iteration of Beers criteria, there have been five subsequent
updated versions; the most recent of which was published in 2019 [4]. Beers crite-
ria, as the longest established explicit IP criteria, are the most widely cited in the
literature. One limitation of Beers criteria is that they do not include instances of
4 Inappropriate Prescription of Medicines 49
PPOs. In contrast, STOPP/START criteria deal both with PIMs and PPOs arranged
by physiological system, as one finds in drug formularies. STOPP/START criteria
were first published in 2008 [5] revised in 2015 [6] and again in 2023 [in press].
There are over 30 other explicit PIM criteria sets in the literature, which describe
a variety of approaches to PIM detection [7]. FORTA criteria, PRISCUS criteria,
French Consensus criteria and NORGEP criteria have all gained significant atten-
tion in the literature. However, Beers criteria and STOPP/START criteria are the
most cited in mainstream online bibliographies. They have also had the most trac-
tion in terms of clinical application in routine practice.
There would be little point in looking for PIMs and PPOs if they were not clinically
relevant or avoidable. However, the literature demonstrates clearly that PIMs and
PPOs are both associated with significant adversity for older people and for the
most part readily detectable and, therefore, preventable. In a prospective study of
600 unselected older patients presenting to hospital with acute illness, Hamilton
et al. showed that ADEs occurred significantly more often in those who were pre-
scribed STOPP criteria-defined PIMs; in contrast, patients taking Beers version 3
criteria-defined PIMs had no significant increase in ADEs [8]. Previous studies had
shown a lack of association between Beers criteria and ADEs [9–11].
The increased risk of ADRs and ADEs arising from PIMs are associated with
several related adverse outcomes for older people, including increased incidence of
acute hospitalization, increased physician consultation, increased healthcare costs
and diminished quality of life [12–14]. Risk factors for PIMs include polypharmacy,
poor functional status and depression [15].
Given the significantly increased risk of ADEs (approximately 85%) when STOPP-
defined PIMs were prescribed to multimorbid older people [8], a series of single-
centre intervention clinical trials were undertaken to test the clinical efficacy of
STOPP/START criteria. These trials are summarized in Table 4.1. They demon-
strated that STOPP/START criteria as an intervention had the following benefits
compared to standard pharmaceutical care:
Table 4.1 Single-centre clinical trials in which STOPP/START criteria were used as an interven-
tion in multimorbid older people with polypharmacy
Target patient
population Impact of intervention compared to usual
Trial authors, year of (number of patients pharmaceutical care (control) group that was
publication randomized) statistically significant
Gallagher et al. [16] Acutely ill Unnecessary polypharmacy, the use of drugs at
hospitalized incorrect doses and potential drug–drug and
multimorbid, aged drug–disease interactions were less frequent in the
(400) intervention group at discharge (absolute risk
reduction 35.7%); underutilization of clinically
indicated medications was also reduced (absolute
risk reduction 21.2%)
Dalleur et al. [17] Hospitalized ‘frail’ Approximately twice as many PIMs removed at
(146) discharge in the intervention group compared to
the control group
Frankenthal et al. [18] Chronic care Fewer drugs, lower care costs and reduced
geriatric facility incidence of falls
residents (359)
O’Connor et al. [19] Acutely ill Reduced ADR incidence in hospital; reduced
hospitalized median monthly medication cost (physician-
multimorbid (732) delivered intervention)
O’Sullivan et al. [20] Acutely ill Reduced ADR incidence (pharmacist-delivered
hospitalized intervention)
multimorbid (737)
Another set of PIM criteria called FORTA (Fit fOR The Aged) criteria devised and
validated by Wehling and colleagues in Germany have also been assessed by clini-
cal trial. FORTA criteria are divided into four categories: A (indispensable), B (ben-
eficial), C (questionable) or D (avoid). In the VALFORTA clinical trial [23], 409
multimorbid patients (cared for in two specialist geriatric medicine units in
Mannheim and Essen) were randomized to a control unit with standard pharmaceu-
tical care or to an intervention unit where a FORTA team instructed attending physi-
cians on FORTA assessment of each prescribed medication. The primary endpoint,
that is, the FORTA score was the sum of medication errors, defined as over-, under-,
or incorrect treatment with prescribed medication. Enrolled patients were random-
ized to intervention or control trial arms in a 1:1 ratio. They had a mean age of
81.5 years; 64% were female, and the mean hospital length of stay was 17.4 days.
The trial results showed a greater reduction of medication errors in the intervention
group versus the control group, with a mean (± SD) FORTA score difference
between admission and discharge in the intervention group of 2.7 (± 2.25) versus
1.0 (± 1.8) in the control group, which was highly significant (p < 0.0001). In addi-
tion, ADRs (not a pre-specified endpoint) were significantly reduced in the interven-
tion group compared to the control group; the absolute risk reduction for incident
ADRs was 19.8%, equating to a number needed to treat of 5 which was once again
statistically significant (p < 0.05). These results are certainly interesting and of
potential clinical importance; however, the VALFORTA trial was a validation trial,
treatment groups not blinded and, to date, results have not been replicated.
intervention group, but not in the basic intervention group, compared to controls.
For the primary composite endpoint, once again patients in the extended interven-
tion group but not those in the basic intervention group experienced a significant
benefit compared to control patients. The hazard ratio (95% CI) for experiencing the
composite primary endpoint was 0.77 (0.64–0.93) representing a number needed to
treat with the extended intervention of 12, that is, a clinically relevant intervention.
The basic pharmacist intervention included structured detailed medication review
with particular focus on aspirin, diuretics, anticoagulants and NSAIDs followed by
a structured report placed in electronic medical record with medication adjustment
recommendations and one-to-one contact with the attending physician. The
extended intervention in the OPTIMIST trial included the basic intervention with
the following elements added:
Table 4.2 Common adverse drug reaction (ADR) presentations in older people in order of fre-
quency (after Jennings et al. [27])
ADR type Frequency Clinical or laboratory manifestation
Fluid and electrolyte 17.3% Hypokalaemia, hyperkalaemia, hyponatraemia,
derangement hypernatraemia, fluid volume overload, fluid volume
depletion
Gastrointestinal motility 14.8% Constipation, diarrhoea
disorders
Renal function 8.2% Acute kidney injury, renal failure complications
impairment
Blood pressure 5.5% Hypotension, orthostatic symptoms
derangement
Delirium 4.1% New onset confusion
Injuries 3.8% Falls
Cardiac arrhythmias 3.7% Bradycardia, atrioventricular block, tachycardia
Other cardiovascular 3.4% Various
disorders
Coagulopathy/bleeding 3.0% Increased INRa, increased APTTb, increased prothrombin
time
Other gastrointestinal 2.8% Various
disorders
Vascular haemorrhagic 2.7% Overt bleeding, haematoma
disorders
Gastrointestinal 2.6% Overt bleeding, malaena
haemorrhage
Neurological disorder 2.5% Various
(unspecified)
Glucose derangement 2.3% Hypoglycaemia, hyperglycaemia
Skin disorder 2.2% Rash, exanthems and other eruptions
Other gastrointestinal 2.2% Nausea/vomiting, dyspepsia, abdominal pain
disorders
a
INR international normalized ratio
b
APTT activated partial thromboplastin time
In the current literature, there are numerous described methods for identifying or
minimizing IP. In a recent review by Rochon et al. [28], a practical method for per-
forming medication review in a way that is designed to minimize IP and its
54 D. O’Mahony
Table 4.3 DRUGS guide to minimizing IP and optimizing medication safety for older adults
(after Rochon et al. [28])
Discuss goals of care and what matters most to the patient
Include patients and caregivers in deprescribing discussions to ensure decisions focus on goals
of care
Review medications
• Encourage patients to bring all prescribed and over-the-counter medications to their
appointment
• Review medications on an ongoing basis and when clinical conditions or goals of care
change
• Discontinue potentially unnecessary drugs
• Consider drug side effects as a potential cause for a new symptom
• Consider non-pharmacological options
• Change for safer alternatives
• Lower the dose
• To identify possible prescribing cascades, determine when the medication was started and
why
Use tools and frameworks
• Identify drugs from the inappropriate prescribing tools, including Beers criteria or STOPP
criteria
• Use the STOPPFrail list when the individual is extremely frail and approaching the end of
life
• Consider whether the new or existing medical condition could be the result of a
prescribing cascadea and ask:
– Is a new drug being prescribed to manage a side effect from another prescribed drug?
– Could the initial drug be replaced with a safer drug or could the dose be reduced?
– Does the patient need the first drug or could this drug be stopped?
– Pay attention to older people who are receiving so-called good drugs with narrow
therapeutic windows that might no longer be needed or for whom dose reduction might be
beneficial
Geriatric medicine approach
Geriatricians carefully consider how multiple medical problems, frailty, cognitive impairment
and limited life expectancy reduce medication benefit, increase adverse events or interfere with
medication adherence
STOP the medications
Consider the algorithm created by Scott [29] or the Good Palliative–Geriatric Practice
algorithm to guide deprescribing [30]
a
‘Prescribing cascades’ refer to inappropriate prescription of drugs for symptoms arising from
side-effects of medications that are misinterpreted as new conditions
consequences has been described. This approach is usefully summarized under the
acronym DRUGS, that is, (1) Discuss goals of care and what matters most to the
patient, (2) Review medications, (3) Use tools and frameworks, (4) Geriatric medi-
cine approach and (5) Stop the medications. More specific details of this approach
are shown in Table 4.3.
Rochon et al. also stressed the importance of patient sex and gender consider-
ations when reviewing and optimizing older people’s medications. Some of these
considerations include the following:
With the rapid growth of the frail older person population aged over 80 years (pre-
dominantly female) in most countries, physicians encounter the challenge of medi-
cation optimization in those older people with a poor 1-year survival prognosis.
These older patients usually have complex multimorbid illness, often have major
impairment of physiological homoeostatic reserve in more than one physiological
system and are usually exposed to high-level polypharmacy. In one study, the asso-
ciation between the extent of chronic multimorbid illness (measured by the
Cumulative Index Rating Scale—Geriatric, CIRS-G) and number of daily prescrip-
tion drugs was a linear one with a high correlation R value of 0.726 [31], i.e. cause-
and-effect is manifest.
Deprescribing in these frailer older people is generally considered desirable in
order to minimize risk of adverse drug–drug and drug–disease interactions as well
as to lessen medication burden for the patients and their carers. Various approaches
to deprescribing have been tried, from implicit criteria algorithms to explicit criteria
for deprescribing of several medication classes, mostly prescribed initially for long-
term disease prevention, in end-of-life situations. In small-scale clinical trials, both
approaches been shown to be both feasible and effective in reducing polypharmacy
and medication burden in multimorbid frailer older people [32–34]. The impact of
deprescribing interventions on clinical outcomes, like incident ADEs, rehospitaliza-
tion and quality of life, however, remains unclear. Approaches to structured depre-
scribing are discussed in greater detail in Chap. 10.
PIMs and PPOs are highly prevalent in older people in all clinical settings. Because
they have clear cut and often serious negative consequences for older people, PIMs
and PPOs should be looked for using appropriate explicit criteria screening tools
56 D. O’Mahony
References
1. Hanlon JT, Schmader KE. The medication appropriateness index at 20: where it started, where
it has been, and where it may be going. Drugs Aging. 2013;30(11):893–900.
2. Bulloch MN, Olin JL. Instruments for evaluating medication use and prescribing in older
adults. J Am Pharm Assoc (2003). 2014;54(5):530–7.
3. Beers MH, Ouslander JG, Rollingher I, Reuben DB, Brooks J, Beck JC. Explicit criteria
for determining inappropriate medication use in nursing home residents. UCLA Division of
Geriatric Medicine. Arch Intern Med. 1991;151(9):1825–32.
4. By the 2019 American Geriatrics Society Beers Criteria® Update Expert Panel. American
Geriatrics Society 2019 updated AGS Beers Criteria® for potentially inappropriate medication
use in older adults. J Am Geriatr Soc. 2019;67(4):674–94.
5. Gallagher P, Ryan C, Byrne S, Kennedy J, O'Mahony D. STOPP (screening tool of older
Person’s prescriptions) and START (screening tool to alert doctors to right treatment).
Consensus validation. Int J Clin Pharmacol Ther. 2008;46(2):72–83.
6. O'Mahony D, O'Sullivan D, Byrne S, O'Connor MN, Ryan C, Gallagher P. STOPP/START
criteria for potentially inappropriate prescribing in older people: version 2. Age Ageing.
2015;44(2):213–8.
4 Inappropriate Prescription of Medicines 57
7. Motter FR, Fritzen JS, Hilmer SN, Paniz ÉV, Paniz VMV. Potentially inappropriate medica-
tion in the elderly: a systematic review of validated explicit criteria. Eur J Clin Pharmacol.
2018;74(6):679–700.
8. Hamilton H, Gallagher P, Ryan C, Byrne S, O'Mahony D. Potentially inappropriate medi-
cations defined by STOPP criteria and the risk of adverse drug events in older hospitalized
patients. Arch Intern Med. 2011;171(11):1013–9.
9. Onder G, Landi F, Liperoti R, Fialova D, Gambassi G, Bernabei R. Impact of inappropriate
drug use among hospitalized older adults. Eur J Clin Pharmacol. 2005;61(5–6):453–9.
10. Laroche ML, Charmes JP, Nouaille Y, Picard N, Merle L. Is inappropriate medication use a
major cause of adverse drug reactions in the elderly? Br J Clin Pharmacol. 2007;63(2):177–86.
11. Budnitz DS, Shehab N, Kegler SR, Richards CL. Medication use leading to emergency depart-
ment visits for adverse drug events in older adults. Ann Intern Med. 2007;147(11):755–65.
12. Davies LE, Spiers G, Kingston A, Todd A, Adamson J, Hanratty B. Adverse outcomes
of polypharmacy in older people: systematic review of reviews. J Am Med Dir Assoc.
2020;21(2):181–7.
13. Xing XX, Zhu C, Liang HY, Wang K, Chu YQ, Zhao LB, Jiang C, Wang YQ, Yan SY. Associations
between potentially inappropriate medications and adverse health outcomes in the elderly: a
systematic review and meta-analysis. Ann Pharmacother. 2019;53(10):1005–19.
14. Hyttinen V, Jyrkkä J, Valtonen H. A systematic review of the impact of potentially inap-
propriate medication on health care utilization and costs among older adults. Med Care.
2016;54(10):950–64.
15. Tommelein E, Mehuys E, Petrovic M, Somers A, Colin P, Boussery K. Potentially inappropri-
ate prescribing in community-dwelling older people across Europe: a systematic literature
review. Eur J Clin Pharmacol. 2015;71(12):1415–27.
16. Gallagher PF, O'Connor MN, O'Mahony D. Prevention of potentially inappropriate prescrib-
ing for elderly patients: a randomized controlled trial using STOPP/START criteria. Clin
Pharmacol Ther. 2011;89(6):845–54.
17. Dalleur O, Boland B, Losseau C, Henrard S, Wouters D, Speybroeck N, Degryse JM, Spinewine
A. Reduction of potentially inappropriate medications using the STOPP criteria in frail older
inpatients: a randomised controlled study. Drugs Aging. 2014;31(4):291–8.
18. Frankenthal D, Lerman Y, Kalendaryev E, Lerman Y. Intervention with the screening tool of
older persons potentially inappropriate prescriptions/screening tool to alert doctors to right
treatment criteria in elderly residents of a chronic geriatric facility: a randomized clinical trial.
J Am Geriatr Soc. 2014;62(9):1658–65.
19. O'Connor MN, O'Sullivan D, Gallagher PF, Eustace J, Byrne S, O'Mahony D. Prevention of
hospital-acquired adverse drug reactions in older people using screening tool of older persons’
prescriptions and screening tool to alert to right treatment criteria: a cluster randomized con-
trolled trial. J Am Geriatr Soc. 2016;64(8):1558–66.
20. O'Sullivan D, O'Mahony D, O'Connor MN, Gallagher P, Gallagher J, Cullinan S, O'Sullivan
R, Eustace J, Byrne S. Prevention of adverse drug reactions in hospitalised older patients using
a software-supported structured pharmacist intervention: a cluster randomised controlled trial.
Drugs Aging. 2016;33(1):63–73.
21. O'Mahony D, Gudmundsson A, Soiza RL, Petrovic M, Jose Cruz-Jentoft A, Cherubini A,
Fordham R, Byrne S, Dahly D, Gallagher P, Lavan A, Curtin D, Dalton K, Cullinan S, Flanagan
E, Shiely F, Samuelsson O, Sverrisdottir A, Subbarayan S, Vandaele L, Meireson E, Montero-
Errasquin B, Rexach-Cano A, Correa Perez A, Lozano-Montoya I, Vélez-Díaz-Pallarés M,
Cerenzia A, Corradi S, Soledad Cotorruelo Ferreiro M, Dimitri F, Marinelli P, Martelli G, Fong
Soe Khioe R, Eustace J. Prevention of adverse drug reactions in hospitalized older patients
with multi-morbidity and polypharmacy: the SENATOR* randomized controlled clinical trial.
Age Ageing. 2020;49(4):605–14.
22. Blum MR, Sallevelt BTGM, Spinewine A, O'Mahony D, Moutzouri E, Feller M, Baumgartner
C, Roumet M, Jungo KT, Schwab N, Bretagne L, Beglinger S, Aubert CE, Wilting I, Thevelin
S, Murphy K, Huibers CJA, Drenth-van Maanen AC, Boland B, Crowley E, Eichenberger A,
Meulendijk M, Jennings E, Adam L, Roos MJ, Gleeson L, Shen Z, Marien S, Meinders AJ,
58 D. O’Mahony
5.1 Introduction
S. A. Sternberg
Department of Geriatric Medicine, Maccabi Healthcare Services, Modiin, Israel
J. H. Gurwitz
Division of Geriatric Medicine and Meyers Primary Care Institute, University of
Massachusetts Medical School, Worcester, MA, USA
e-mail: [email protected]
P. A. Rochon (*)
Women’s Age Lab, Women’s College Hospital, Toronto, ON, Canada
Department of Medicine, University of Toronto, Toronto, ON, Canada
Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health,
University of Toronto, Toronto, ON, Canada
e-mail: [email protected]
Prescribing cascades were described by Rochon and Gurwitz in the 1990s [1, 2].
One typical example of a prescribing cascade is a non-steroidal anti-inflamma-
tory drug (NSAID) leading to hypertension and subsequently leading to the ini-
tiation of a new anti-hypertensive medication prescription [2]. This definition
was expanded in 2017 [3] to include the prescribing of potentially unnecessary
over-the-counter drug therapies and medical devices such as permanent pace-
maker devices to counteract severe bradyarrhythmias unrecognized as being
caused by cardiovascular drugs like beta-blockers, digoxin and verapamil.
Prescribing cascades must, by definition, reflect inappropriate prescribing
(Fig. 5.1).
Older adults are particularly at risk for prescribing cascades and drug-related harm
due to the high prevalence of multimorbidity and associated polypharmacy. Most
older adults have multiple medical conditions and drug therapies that are used to
treat these conditions [11]. Polypharmacy is also extremely common in older adults,
especially in the long-term care setting [12]. In the United States, the use of five or
more medications by older adults over age 65 has increased from 13% in 1988 to
39% in 2010 [13]. The prevalence of polypharmacy in European older adults ranged
from 26% in Switzerland to 40% in the Czech Republic, as reported by the Survey
of Health, Ageing and Retirement in Europe (SHARE) based on 2015 data from 17
countries [14]. Among older adults living in long-term care in Europe and Israel,
almost half used between five and nine daily drug therapies, and almost a quarter
used ten or more drugs [15]. For older adults living in long-term care homes in
Canada, where almost 70% of residents are women [16], almost 50% were pre-
scribed medications from ten or more drug classes [12]. This high rate of hyperpoly-
pharmacy (i.e. ten or more daily drugs) is related not only to multimorbidity but is
also a consequence of multiple prescribers caring for older people without adequate
communication between the prescribers. In addition, clinical guidelines tend to
focus on single diseases without taking into consideration that two-thirds of adults
over the age of 65 have multiple chronic conditions requiring the use of numerous
medications [11]. Finally, current prescribing guidelines do not routinely take into
consideration an older adult’s goals of care or their remaining life expectancy [17].
Important factors such as a person’s cognitive and frailty status are insufficiently
considered when assessing the potential risks and benefits of a medication [18]. The
combination of these factors increases the risk of prescribing cascades occurring in
older adults.
Among older adults, prescribing cascades may disproportionately impact
older women. Older women have the most chronic conditions and often use the
greatest number of drug therapies [19]. In addition, sex (female biology) and
gender (sociocultural construct), in combination, contribute to this increased risk
of prescribing cascades. The pharmacokinetic and pharmacodynamic changes
associated with the female sex and older age contribute to the increased preva-
lence of adverse drug events in women compared to men [20–23]. Furthermore,
it was found that older European women were more likely than men to have
multimorbidity and as a result were more likely to be prescribed multiple
62 S. A. Sternberg et al.
In 1997, three initial prescribing cascades were described [2]. The original exam-
ples were: NSAIDs leading to a prostaglandin-mediated rise in blood pressure with
the subsequent prescription of antihypertensive therapy in older people; thiazide
diuretics prescribed to treat hypertension leading to hyperuricemia and the subse-
quent prescription of colchicine for gout; and the initiation of antipsychotic drug
therapy followed by the development of Parkinsonism, which was misinterpreted as
Parkinson’s disease and treated with levodopa/carbidopa. As awareness has
increased, so have the number of prescribing cascades. In 2017 [3], there were 20
examples of prescribing cascades, and recently, more than 100 prescribing cascades
have been identified as part of an international project [7]. Examples of prescribing
cascades can be seen in Table 5.1.
One notable example of a prescribing cascade is a cholinesterase inhibitor (e.g.
donepezil) prescribed to manage the symptoms of dementia leading to the develop-
ment of urinary incontinence with the subsequent initiation of a urinary anticholin-
ergic drug to treat the incontinence [6]. The concern about starting a urinary
anticholinergic agent is that this therapy can lead to additional side effects, includ-
ing dry mouth, constipation and confusion. Another example of a prescribing cas-
cade is a calcium channel blocker (CCB) leading to ankle oedema and the subsequent
prescribing of loop diuretics [4]. This cascade was described in a large population-
based cohort study that included 41,000 adults over age 65 (59% women) who were
newly prescribed a CCB. After adjustment, individuals who were newly dispensed
a CCB had significantly increased relative rates of being dispensed a loop diuretic
compared with individuals who were newly dispensed an angiotensin-converting
enzyme inhibitor or angiotensin II receptor blocker. Given that calcium channel
blockers are among the top ten prescribed medications, and that 2–25% of CCBs
5 Prescribing Cascades 63
cause peripheral oedema, this prescribing cascade has the potential to cause impor-
tant drug-related harm to older adults.
Another population-based study investigated the prescribing cascade of sodium
glucose cotransporter 2 inhibitor (SGLT2) medications leading to genital fungal
infection and the subsequent initiation of an anti-fungal medication to treat this
SGLT2 side effect [29]. It compared 21,444 new users of SGLT2 inhibitors to
22,463 incident users of dipeptidyl-peptidase-4 (DPP4) inhibitors. After adjusting
for propensity score, age, sex and recent urinary tract infection, there was a 2.47-
fold increased risk of genital fungal infection (determined by the use of antimycotic
medication) with incident use of SGLT2 inhibitors within 30 days compared to
incident use of DPP4 inhibitors. This prescribing cascade should be an important
consideration when using SGLT2 medications for older adults with diabetes and
cardiovascular disease.
In the expanded version of the prescribing cascade definition, over-the-counter
(OTC) drugs and medical devices were also included (Table 5.2). One case study
64 S. A. Sternberg et al.
Table 5.2 Examples of the expanded definition of prescribing cascades involving over-the-
counter therapies and medical devices
Over-the-counter therapy
Initial drug therapy Adverse event misinterpreted as a Subsequent over-the-counter
prescribed new medical condition drug therapy
Cholinesterase inhibitor Gastrointestinal symptoms Bismuth subsalicylate
[30]
Cholinesterase inhibitor Rhinorrhoea Diphenhydramine
[43]
Enalapril [36] Cough Dextromethorphan
Bupropion [44] Insomnia Sleep medication
Ramipril [44] Cough Cough syrup
Medical device
Initial drug therapy Adverse event misinterpreted as a Medical device
prescribed new medical condition
Cholinesterase Bradycardia Pacemaker device
inhibitorsa [45]
a
The article did not mention the term prescribing cascade in the text, but described the concept of
a prescribing cascade
[30] example involved an acetylcholinesterase that was prescribed for dementia and
subsequently led to abdominal discomfort. The patient self-medicated with large
quantities of bismuth salicylate causing him to suffer from salicylate toxicity and
bismuth neurotoxicity, leading to a fall and hospitalization for subdural haemor-
rhage and bilateral frontal contusions. This case illustrates that prescribing cascades
can occur with OTC drugs and with prescription medications. In addition, prescrib-
ing cascades can increase the risk of serious consequences such as morbidity and
hospitalization.
Identifying and preventing prescribing cascades can inform better clinical care.
Many tools and frameworks have been described to identify inappropriate prescrib-
ing [11] including the prescribing cascade framework. Providing a detailed medica-
tion history, particularly when a medication was started, would help clinicians
determine the drug prescribing sequence and facilitate the early recognition of a
prescribing cascade. One such tool includes four simple questions to help physi-
cians identify and interrupt prescribing cascades [10]. These questions were devel-
oped to raise awareness of prescribing cascades each time a new drug is prescribed.
Providers need to think of prescribing cascades and be adept at identifying when
they occur, in order to prevent them (Box 5.1).
5 Prescribing Cascades 65
Physicians are often confronted with prescribing cascades in their clinical prac-
tice. For example, enalapril is prescribed for hypertension leading to a persistent dry
cough and an antibiotic such as levofloxocin is prescribed for suspected underlying
respiratory infection. Atenolol, a beta-blocker, is prescribed after a cardiac event,
leading to depression, and escitalopram is prescribed to treat the depression.
Table 5.1 presents examples of drug classes and specific examples of prescribing
cascades reported by physicians in clinical practice, and Table 5.2 provides some
examples of prescribing cascades involving OTC medications and medical devices,
as described in the expanded definition (see Fig. 5.1). Prescribing cascades have
also been incorporated into many deprescribing protocols [11]. Deprescribing is the
process of tapering or stopping medications that may not be indicated, in accor-
dance with the patients’ preferences, to minimize polypharmacy and improve
patient outcomes [31]. Most deprescribing protocols review all medications and
indications for use, consider each drugs risk/benefit profile and then develop a pri-
oritized plan for deprescribing. The medication review stage provides an opportu-
nity to identify prescribing cascades and then determine their potential for
deprescribing. Process mapping is another tool that can be used by clinicians to
identify prescribing cascades in their patients [32]. A clinical process map is a tool
designed to graphically illustrate a patient’s presentation, symptoms, medications,
possible side effects and interventions. This is generated quickly at the bedside by
the clinician to help visualize the sequence of events leading to the development of
the prescribing cascade and to understand its consequences.
5.6 Conclusions
Prescribing cascades are well documented in the literature and are an important
cause of drug-related harm in older adults. Older women are at higher risk for drug-
related harm than older men. Prescribing cascades can be prevented by implement-
ing interventions at each stage of drug prescription and use. At the time of
prescribing, the provider should be aware of potential prescribing cascades that may
arise from the drug being initiated and carefully document the date that each new
drug was started. At regular follow-up, providers should question whether a new
complaint could be a medication side effect and whether a new prescription is really
necessary. At medication review, validated tools and frameworks should be applied
66 S. A. Sternberg et al.
References
1. Rochon PA, Gurwitz JH. Drug therapy. Lancet. 1995;346(8966):32–6.
2. Rochon PA, Gurwitz JH. Optimising drug treatment for elderly people: the prescribing cas-
cade. BMJ. 1997;315(7115):1096–9.
3. Rochon PA, Gurwitz JH. The prescribing cascade revisited. Lancet. 2017;389(10081):1778–80.
4. Savage RD, Visentin JD, Bronskill SE, et al. Evaluation of a common prescribing cascade of
calcium channel blockers and diuretics in older adults with hypertension. JAMA Intern Med.
2020;180(5):643–51.
5. Read SH, Giannakeas V, Pop P, et al. Evidence of a gabapentinoid and diuretic prescribing
cascade among older adults with lower back pain. J Am Geriatr Soc. 2021;69(10):2842–50.
6. Gill SS, Mamdani M, Naglie G, et al. A prescribing cascade involving cholinesterase inhibitors
and anticholinergic drugs. Arch Intern Med. 2005;165(7):808–13.
7. Sternberg SA, Petrovic M, Onder G, et al. Identifying key prescribing cascades in older people
(iKascade): a transnational initiative on drug safety through a sex and gender lens—rationale
and design. Eur Geriatr Med. 2021;12(3):475–83.
8. Choosing Wisely Australia. 5 Things clinicians and consumers should question. Developed
by the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists.
2016. http://www.choosingwisely.org.au/recommendations/ascept. Accessed 9 July 2021.
9. Onder G, Bonassi S, Abbatecola AM, et al. High prevalence of poor quality drug prescribing in
older individuals: a nationwide report from the Italian Medicines Agency (AIFA). J Gerontol
A Biol Sci Med Sci. 2014;69(4):430–7.
10. Sternberg SA, Guy-Alfandary S, Rochon PA. Prescribing cascades in older adults.
CMAJ. 2021;193(6):E215.
11. Rochon PA, Petrovic M, Cherubini A, et al. Polypharmacy, inappropriate prescrib-
ing, and deprescribing in old age:through a sex and gender lens. Lancet Healthy Longev.
2021;2(5):E290–300.
12. Canadian Institute for Health Information. Drug use among seniors in Canada, 2016. Ottawa:
CIHI; 2018. https://secure.cihi.ca/free_products/drug-use-among-seniors-2016-en-web.pdf.
Accessed 9 July 2021
13. Guthrie B, Makubate B, Hernandez-Santiago V, Dreischulte T. The rising tide of polypharmacy
and drug–drug interactions: population database analysis 1995–2010. BMC Med. 2015;13:74.
14. Midao L, Giardini A, Menditto E, Kardas P, Costa E. Polypharmacy prevalence among older
adults based on the survey of health, ageing and retirement in Europe. Arch Gerontol Geriatr.
2018;78:213–20.
15. Onder G, Liperoti R, Fialova D, et al. Polypharmacy in nursing home in Europe: results from
the SHELTER study. J Gerontol A Biol Sci Med Sci. 2012;67(6):698–704.
16. Statistics Canada. 2016 Census of population, statistics Canada catalogue no. 98-400-
X2016018. 2019. https://www12.statcan.gc.ca/census-recensement/2016/geo/geosearch-
georecherche/ips/index.cfm?q=98-400-X2016018&l=en&g=2016A000011124. Accessed 9
Sept 2021.
17. Stall NM, Fischer HD, Fung K, et al. Sex-specific differences in end-of-life burdensome inter-
ventions and antibiotic therapy in nursing home residents with advanced dementia. JAMA
Netw Open. 2019;2(8):e199557.
18. Holmes HM, Hayley DC, Alexander GC, Sachs GA. Reconsidering medication appropriate-
ness for patients late in life. Arch Intern Med. 2006;166(6):605–9.
5 Prescribing Cascades 67
19. Gurwitz JH. The age/gender interface in geriatric pharmacotherapy. J Womens Health
(Larchmt). 2005;14(1):68–72.
20. Watson S, Caster O, Rochon PA, den Ruijter H. Reported adverse drug reactions in women
and men: aggregated evidence from globally collected individual case reports during half a
century. EClinicalMedicine. 2019;17:100188.
21. Alturki A, Alaama T, Alomran Y, Al-Jedai A, Almudaiheem H, Watfa G. Potentially inappro-
priate medications in older patients based on beers criteria: a cross-sectional study of a family
medicine practice in Saudi Arabia. BJGP Open. 2020;4(1):bjgpopen20X101009.
22. Rydberg DM, Mejyr S, Loikas D, Schenck-Gustafsson K, von Euler M, Malmström RE. Sex
differences in spontaneous reports on adverse drug events for common antihypertensive drugs.
Eur J Clin Pharmacol. 2018;74(9):1165–73.
23. Sørup FKH, Eriksson R, Westergaard D, Hallas J, Brunak S, Ejdrup AS. Sex differences in text-
mined possible adverse drug events associated with drugs for psychosis. J Psychopharmacol
(Oxford, England). 2020;34(5):532–9.
24. Bezerra de Souza DL, Oliveras-Fabregas A, Espelt A, et al. Multimorbidity and its associated
factors among adults aged 50 and over: a cross-sectional study in 17 European countries. PLoS
One. 2021;16(2):e0246623.
25. McCormick A, Charlton J. Morbidity statistics from general practice. In: Fourth national study
1991–1992. OPCS series MB5 no. 3. London: HMSO; 1995.
26. Cheff R, Hill M, Iveniuk J. Who benefits? Gaps in medication coverage for Ontario work-
ers. Toronto: Wellesley Institute; 2019. https://www.wellesleyinstitute.com/wp-content/
uploads/2019/12/Coverage-Gaps-for-Ontario-Workers_EMBARGO_27.11.19.pdf. Accessed
9 July 2021
27. Mauvais-Jarvis F, Bairey Merz N, Barnes PJ, et al. Sex and gender: modifiers of health, dis-
ease, and medicine. Lancet (London, England). 2020;396(10250):565–82.
28. Regitz-Zagrosek V. Sex and gender differences in pharmacology. Berlin: Springer; 2012.
29. Lega IC, Bronskill SE, Campitelli MA, et al. Sodium glucose cotransporter 2 inhibitors and
risk of genital mycotic and urinary tract infection: a population-based study of older women
and men with diabetes. Diabetes Obes Metab. 2019;21(11):2394–404.
30. Rosenberg J, Rochon PA, Gill SS. Unveiling a prescribing cascade in an older man. J Am
Geriatr Soc. 2014;62(3):580–1.
31. Frank C, Weir E. Deprescribing for older patients. CMAJ. 2014;186(18):1369–76.
32. Piggott KL, Mehta N, Wong CL, Rochon PA. Using a clinical process map to identify prescrib-
ing cascades in your patient. BMJ. 2020;368:m261.
33. Kalisch LM, Caughey GE, Roughead EE, Gilbert AL. The prescribing cascade. Aust Prescr.
2011;34(6):162–6.
34. Liu PT, Argento VS, Skudlarska BA. Prescribing cascade in an 80-year-old Japanese immi-
grant. Geriatr Gerontol Int. 2009;9(4):402–4.
35. Rababa M, Al-Ghassani AA, Kovach CR, Dyer EM. Proton pump inhibitors and the prescrib-
ing cascade. J Gerontol Nurs. 2016;42(4):23–31.
36. Kwan D, Farrell B. Polypharmcy: optimizing medication use in elderly patients. CGS J
CME. 2014;4(1):21–7.
37. Rochon PA, Stukel TA, Sykora K, et al. Atypical antipsychotics and Parkinsonism. Arch Intern
Med. 2005;165(16):1882–8.
38. Singh S, Cocoros NM, Haynes K, et al. Antidopaminergic–antiparkinsonian medication pre-
scribing cascade in persons with Alzheimer’s disease. J Am Geriatr Soc. 2021;69(5):1328–33.
39. Singh S, Cocoros NM, Haynes K, et al. Identifying prescribing cascades in Alzheimer’s
disease and related dementias: the calcium channel blocker-diuretic prescribing cascade.
Pharmacoepidemiol Drug Saf. 2021;30(8):1066–73.
40. Trenaman SC, Bowles SK, Kirkland S, Andrew MK. An examination of three prescribing
cascades in a cohort of older adults with dementia. BMC Geriatr. 2021;21(1):297.
41. Vouri SM, van Tuyl JS, Olsen MA, Xian H, Schootman M. An evaluation of a potential cal-
cium channel blocker-lower-extremity edema-loop diuretic prescribing cascade. J Am Pharm
Assoc (2003). 2018;58(5):534–9 e4.
68 S. A. Sternberg et al.
42. Ponte ML, Wachs L, Wachs A, Serra HA. Prescribing cascade. A proposed new way to evalu-
ate it. Medicina (B Aires). 2017;77(1):13–6.
43. Vouri SM, Chung JM, Binder EF. Successful intervention to mitigate an acetylcholines-
terase inhibitor-induced rhinorrhea prescribing cascade: a case report. J Clin Pharm Ther.
2017;42(3):370–1.
44. Gnon C, Currie J, Trimble J. Are you the victim of a prescribing cascades? Candian depre-
scribing network. 2020. https://www.deprescribingnetwork.ca/blog/prescribing-cascade.
Accessed 28 Sept 2021.
45. Gill SS, Anderson GM, Fischer HD, et al. Syncope and its consequences in patients with
dementia receiving cholinesterase inhibitors: a population-based cohort study. Arch Intern
Med. 2009;169(9):867–73.
Adverse Drug Reactions in Older People:
A Twenty-First Century View 6
Mary Randles and Denis O’Mahony
6.1 Introduction
The unprecedented number of adults living beyond 65 years is one of the great suc-
cess stories of modern civilisation and modern medicine. Over the past century, the
proportion of older adults in the United States of America has increased threefold,
similar to like western Europe and most developed countries globally, leading to an
increase in life expectancy that has not yet reached a plateau [1, 2]. The proportion
of adults over 65 years in the United States was recently calculated at 20.3%, repre-
senting an increase of 2.9% over the 10 years to 2019. While these increases in
longevity represent progress in relation to societal health in general, they also pose
many challenges currently and in the future in terms of growing levels of comorbid-
ity and frailty accompanied by higher prevalence of polypharmacy and adverse drug
reactions in the population of any country observing societal ageing.
The success of medical management and pharmacotherapy in this population is
not straightforward. It is generally accepted that medical interventions have facili-
tated older people to increase their longevity and maintain generally better health.
However, greater exposure to polypharmacy has resulted in an ever-increasing
potential for adverse drug reactions (ADRs), adverse drug events (ADEs) and
adverse drug withdrawal events (ADWEs).
An adverse drug reaction was originally defined by the World Health Organisation
(WHO) over 50 years ago as: “A response to a drug that is noxious and unintended
and occurs at doses normally used in man for the prophylaxis, diagnosis or therapy
of disease, or for the modification of physiological function” [3]. This WHO defini-
tion was developed with the aim of monitoring ADRs, particularly in drug develop-
ment trials and soon after approval of drugs for marketing for general use. Edwards
and Aronson further defined an ADR as: “An appreciably harmful or unpleasant
reaction, resulting from an intervention related to the use of a medicinal product,
which predicts hazard from future administration and warrants prevention or spe-
cific treatment, or the alteration of the dosage regimen, or withdrawal of the prod-
uct” [4]. They further point out that while an adverse drug effect and adverse drug
reaction are often used interchangeably, an adverse drug effect is seen from the
point of view of the drug, whereas the adverse drug reaction is seen from the point
of view of the patient [4]. Edwards and Aronson’s definition was further expanded
in 2010 to include reactions occurring as a result of error, misuse or abuse and to
suspected reactions to medicines that are unlicensed or used off label in addition to
the authorised use of a medicinal product in normal doses [5].
When discussing ADRs, some experts emphasise the need to differentiate
between ADRs and ADEs. The difference depends essentially on the adverse effect.
While we can define an ADE as an adverse outcome that occurs while a patient is
taking a drug, it is not necessarily directly attributable to the drug [4], such as the
increased incidence of falls and fractures when older people are taking
benzodiazepines.
While historically there has been little interest in the specific effect of ADRs in
older adults, over the past two decades, there has been increasing focus on the prev-
alence, incidence, clinical effect and cost of ADRs in older adults. In the general
adult hospitalised population, ADRs account for approximately 6% of all admis-
sions [6]. Furthermore, prospective analysis of inpatient episodes demonstrated that
14.7% (95% CI, 13.6–15.9%) of patients experienced at least one incident ADR [6,
7]. Given the increased complexity of comorbid diseases and polypharmacy in older
people, it is unsurprising that a systematic review and meta-analysis [8] found the
prevalence of ADRs in older adults in acute care settings was greater than in the
general adult population. The overall mean prevalence of ADRs in the same review
was 11.0% (95% CI: 5.1–16.8) with 11.5% occurring during hospitalisation. This
meta-analysis demonstrated a wide variation of overall ADR prevalence across the
included studies, that is, from 5.8% [8] to 46.3% [9]. The authors note that prospec-
tive studies and larger scale studies generally found higher ADR prevalence rates
[10] in studies of ADRs in ‘old old’ adults (i.e. over 80 years), reporting an average
in-hospital ADR incidence of 13%. Although the large variance in the prevalence of
6 Adverse Drug Reactions in Older People: A Twenty-First Century View 71
ADRs observed in older hospitalised adults has been attributed to the difference in
sample sizes and different admission settings within hospitals (general unselected
medical acute admissions versus dedicated geriatric medicine unit admissions) [11],
it remains very clear that ADRs are a common problem faced by all clinicians treat-
ing older adults.
Highly variable prevalence rates of ADRs have been reported in community-
dwelling older adults also, varying from 6.5% in one prospective study [6] to 78%
in a retrospective cohort of older adults where the ADRs were self-reported by
patients and corroborated by clinician review [12]; the latter study also included
drug events that may not have resulted in the index hospitalisation. Older adults in
long-term care settings were found to have an ADR prevalence of 14% with other
studies reporting an ADR incidence in long-term care older patients between 1.19
and 7.26 per 100 resident-months [12]. It is estimated that older adults are approxi-
mately seven times more likely to experience an ADR requiring hospital admission,
and when hospitalised they are substantially more likely to experience ADRs during
their hospital admission than younger patients [13, 14].
ADRs in older adults result in excess economic costs during hospital admissions,
increased length of stay, additional cost of care and increased mortality. A French
study evaluating the cost of ADRs with the aim of assessing the additional financial
resource utilisation arising from ADRs found that the ADR incidence increased
with age and that half of the ADRs led to additional investigations and increased
median length of stay, that is, 11 days versus 7 days in those patients who did not
experience ADRs. This increase in length of stay resulted in a mean cost of approxi-
mately €1800 per ADR [15], similar to the cost of €2128 per ADR-related hospitali-
sation calculated by Beijer and Blaey [16]. ADRs have a significant impact on
hospital running costs accounting for 5% of overall hospital running costs in the
USA, estimated at between $3.5 billion and $30 billion per annum [13–15, 17]. The
fatality rate from ADRs has been reported as 0.15% in a larger scale, retrospective,
population-based study and 0.44% in older adults in one meta-analysis [6, 18].
Metabolism of drugs may be altered with ageing also due to reductions in hepatic
volume, renal and hepatic perfusion, and protein binding. Older people who experi-
ence polypharmacy are more likely to take drugs that cause inhibition or activation
of the CYP pathway [11, 20]. Normal physiological ageing and common disorders
such as cardiovascular disease, diabetes, heart failure and hypertension can all lead
to reduced renal clearance and excretion of drugs [20].
Multimorbidity, the co-occurrence of two or more coinciding medical conditions
was shown to affect up to 95.1% of patients aged over 65 in the primary care setting
in a systematic review of 39 studies [21]. Older people are at considerably higher risk
of experiencing polypharmacy due to a much higher burden of multimorbidity than is
encountered in younger people. Polypharmacy, as the principal risk factor for ADRs,
may be engendered and exacerbated by older people attending multiple specialists for
individual diseases or conditions, leading to more evidence-based drug therapies.
In one study [22], researchers found that taking five or more daily medications
was associated with an odds ratio for ADEs of 1.88 (95% CI, 1.58–2.25) compared
to patients taking zero to four daily medications). Over the last 20 years, there has
been a notable increase in treatment guidelines for many common conditions, par-
ticularly in cardiovascular disease and diabetes where treatment guidelines recom-
mend multiple drugs for secondary prevention and ongoing management of these
conditions. Worryingly, studies demonstrate that polypharmacy has increased in the
last decade, particularly after hospital admissions [23, 24]. Higher levels of poly-
pharmacy increase the risk of both drug–disease interactions and drug–drug interac-
tions [25].
Although older patients experience most of the burden of multimorbidity and poly-
pharmacy and are the largest group of patients receiving evidence-based pharmaco-
therapies, paradoxically, they are often excluded from clinical drug trials because of
their multimorbidity. An example of this discrepancy can be seen in cancer drug trials.
While people over 65 years represent over 60% of new cancer cases in Europe and the
US annually, they only account for 22% of patients recruited into cancer trials; this
proportion drops to 8–13% in those aged over 70 years [26]. This degree of older
patient exclusion from clinical trials is further illustrated in a review of 440 trials of
drugs for type 2 diabetes [27]. Despite being the most common long-term metabolic
condition experienced by older adults, only six trials were specifically designed to
study older adults; 289 trials excluded individuals based on an arbitrary age cut-off
and a further 76.8% of trials excluded people with comorbidities.
Historically, ADRs were classified as Type A and Type B reactions. In Rawlins and
Thompson’s classification, Type A reactions were typically associated with drug
doses and were predictable from the pharmacological properties and physiological
actions of the drug. Type B or “Bizarre” reactions were not seen as dose related,
such as idiosyncratic hypersensitivity reactions [28]. Over time, these categorical
ADR types were expanded to include further four categories; (Table 6.1) Type C
6
of drug benzodiazepines
• Myocardial ischemia after
withdrawal of beta-blockers
F Failure Yes Unexpected failure of therapy • Inadequate anticoagulation • Increase in dosage
• Dose related effect from warfarin with • Review of effects of
• Drug interaction related carbamazipine concomitant therapy
(Adapted from Edwards & Aronson, 2000)
73
74 M. Randles and D. O’Mahony
Table 6.2 Types of time-dependent drug reactions based of Aronson and Ferner ADR classifica-
tion [29]
Time-dependant reactions
Type of
reaction Features Example
Rapid reactions Occur only when a drug is administered Vancomycin → red man syndrome
too quickly
First dose Occur after the first dose of a certain Ace inhibitor-hypotension
reactions treatment. May or may not occur again Type I hypersensitivity reaction to
penicillin
Early reactions Occur early in treatment but abate as Acetylcholinesterase inhibitors →
treatment progresses gastrointestinal disturbance or
nausea
Intermediate Occur after some delay, however if a Type II–IV hypersensitivity
reactions reaction hasn’t occurred after a certain reactions
time, the population risk falls
Late reactions Risk increases with repeated exposure Dopamine receptor antagonists →
Also includes withdrawal reactions tardive dyskinesias
Corticosteroids → osteoporosis
Delayed Effects are observed at some time after Carcinogens
reactions exposure, even if the drug has been
withdrawn
6 Adverse Drug Reactions in Older People: A Twenty-First Century View 75
Table 6.3 Adverse drug reactions (ADR) causality matrix adapted from World Health
Organisation–Upsala Monitoring Centre (WHO–UMC) ADR causality criteria [36]
Categories of Time sequence Other drugs/ Dechallenge Rechallenge
ADR consistent with diseases ruled response, that is, response, that is,
attribution ADR out as causative clinical improvement clinical relapse
Certain Yes Yes Yes Yes
Probable Yes Yes Yes No
Possible Yes No No No
Unlikely No No No No
76 M. Randles and D. O’Mahony
Table 6.4 Hartwig adverse drug reactions (ADR) severity scale and Hallas ADR avoidability
criteria
Hartwig ADR severity grade (Hartwig Hallas ADR avoidability criteria (Hallas et al. 1990)
et al. 1992) [37] [38]
1 An ADR occurred but no Definitely The drug event was due to a drug
change in treatment with avoidable treatment procedure inconsistent with
suspected drug present-day knowledge of good
medical practice or was clearly
unrealistic, taking the known
circumstances into account
2 The ADR required that Possibly The prescription was not erroneous,
treatment with the suspected avoidable but the drug event could have been
drug be held, discontinued or avoided by an effort exceeding the
otherwise changed (no antidote obligatory demands
required and no increase in
length of stay)
3 The ADR required that Unavoidable The drug event could not have been
treatment with the suspected avoided by any reasonable means, or
drug be held, discontinued or it was an unpredictable event in the
otherwise changed or and an course of a treatment fully in
antidote or other treatment was accordance with good medical
required practice
4 Any level 3 ADR which Unclassifiable The data for rating could not be
resulted in admission or obtained or the evidence was
increased length of stay by at conflicting
least 1 day
5 Any level 4 ADR which
required intensive medical care
6 Any ADR causing permanent
harm to the patient
7a The ADR indirectly linked to
death in the patient
7b The ADR was directly linked to
death in the patient
clinicians caring for older patients need to maintain a heightened vigilance for
detection of possible ADRs and to carefully examine all presentations to ensure that
ADRs are not overlooked as a diagnosis.
ADRs are a potentially preventable form of iatrogenic harm. In the past decade,
interest in developing new clinical tools designed to reduce and prevent ADRs in
older people has grown. Given the association between inappropriate prescribing
(IP) and ADRs, explicit IP assessment tools such as STOPP/START criteria and
Beers criteria, although developed to identify and reduce IP, may have potential to
reduce ADRs. In one single-centre, single-blinded study, application of STOPP/
START criteria (version 1) to the medication lists of acutely hospitalised older
6 Adverse Drug Reactions in Older People: A Twenty-First Century View 77
Table 6.5 The ten most common drug classes causing adverse drug reactions (ADRs) in hospital-
ised older patients [43]
Drug class (in rank order of Proportion of total Most commonly reported
frequency) reported ADRs pharmacological subgroup
Diuretics 19.83% Loop (high ceiling) diuretics
Antibacterials (systemic 14.84% Beta-lactams (penicillins,
use) cephalosporins), macrolides, quinolones
Antithrombotics 12.24% VKAsa, heparin and heparinoids,
antiplatelet agents
Analgesics 10.90% Opioids
Drugs for obstructive 4.74% Beta-2 adrenergic agonists, xanthines
airways disease (e.g. theophylline)
Renin–angiotensin system 4.11% ACE inhibitors, angiotensin-II receptor
antagonists blockers
Psychotropics 3.86% Benzodiazepines, antipsychotics
Corticosteroids (systemic 3.23% Glucocorticoids
use)
Cardiac drugs 2.98% Cardiac glycosides, vasodilators, class I
and III antiarrhythmics
Antidiabetic drugs 2.56% Insulin and insulin analogues, oral
antidiabetics
Vitamin K antagonists, for example, warfarin
a
78 M. Randles and D. O’Mahony
6.8 Conclusion
References
1. Eurostat. Population structure and aging 2020. https://ec.europa.eu/eurostat/statistics-
explained/index.php/Population_structure_and_ageing#Past_and_future_population_age-
ing_trends_in_the_EU-27.
2. Anderson LA, Goodman RA, Holtzman D, Posner SF, Northridge ME. Aging in the United
States: opportunities and challenges for public health. Am J Public Health. 2012;102(3):393–5.
3. World Health Organization. International drug monitoring: the role of national centres, report
of a WHO meeting [held in Geneva from 20 to 25 September 1971]. Geneva: World Health
Organization; 1972.
4. Edwards IR, Aronson JK. Adverse drug reactions: definitions, diagnosis, and management.
Lancet. 2000;356(9237):1255–9.
5. Parliament E, December Co. Directive 2010/84/EU of The European Parliament and of the
Council of 15 December 2010, amending, as regards to pharmacovigilance, Directive 2001/83/
EC on the Community Code relating to medicinal products for human use. 2010.
6 Adverse Drug Reactions in Older People: A Twenty-First Century View 79
6. Pirmohamed M, James S, Meakin S, Green C, Scott AK, Walley TJ, et al. Adverse drug
reactions as cause of admission to hospital: prospective analysis of 18,820 patients.
BMJ. 2004;329(7456):15–9.
7. Davies EC, Green CF, Taylor S, Williamson PR, Mottram DR, Pirmohamed M. Adverse drug
reactions in hospital in-patients: a prospective analysis of 3695 patient-episodes. PLoS One.
2009;4(2):e4439.
8. Franceschi M, Scarcelli C, Niro V, Seripa D, Pazienza AM, Pepe G, et al. Prevalence, clinical
features and avoidability of adverse drug reactions as cause of admission to a geriatric unit.
Drug Saf. 2008;31(6):545–56.
9. De Paepe P, Petrovic M, Outtier L, Van Maele G, Buylaert W. Drug interactions and adverse
drug reactions in the older patients admitted to the emergency department. Acta Clin Belg.
2013;68(1):15–21.
10. Alhawassi TM, Krass I, Bajorek BV, Pont LG. A systematic review of the prevalence and risk
factors for adverse drug reactions in the elderly in the acute care setting. Clin Interv Aging.
2014;9:2079–86.
11. Davies EA, O'Mahony MS. Adverse drug reactions in special populations—the elderly. Br J
Clin Pharmacol. 2015;80(4):796–807.
12. Handler SM, Wright RM, Ruby CM, Hanlon JT. Epidemiology of medication-related adverse
events in nursing homes. Am J Geriatr Pharmacother. 2006;4(3):264–72.
13. Tangiisuran B, Gozzoli M, Davies J, Rajkumar C. Adverse drug reactions in older people. Rev
Clin Gerontol. 2010;20(3):246.
14. White TJ, Arakelian A, Rho JP. Counting the costs of drug-related adverse events.
PharmacoEconomics. 1999;15(5):445–58.
15. Patel K, Kedia M, Bajpai D, Mehta S, Kshirsagar N, Gogtay N. Evaluation of the prevalence
and economic burden of adverse drug reactions presenting to the medical emergency depart-
ment of a tertiary referral centre: a prospective study. BMC Clin Pharmacol. 2007;7(1):1–5.
16. Beijer H, De Blaey C. Hospitalisations caused by adverse drug reactions (ADR): a meta-
analysis of observational studies. Pharm World Sci. 2002;24(2):46–54.
17. Cronenwett LR, Bootman JL, Wolcott J, Aspden P. Preventing medication errors. Washington,
DC: National Academies Press; 2007.
18. Patel TK, Patel PB. Mortality among patients due to adverse drug reactions that lead to hospi-
talization: a meta-analysis. Eur J Clin Pharmacol. 2018;74(6):819–32.
19. Corsonello A, Pedone C, Incalzi RA. Age-related pharmacokinetic and pharmacodynamic
changes and related risk of adverse drug reactions. Curr Med Chem. 2010;17(6):571–84.
20. Corsonello A, Pedone C, Lattanzio F, Onder G, Incalzi RA. Association between glomeru-
lar filtration rate and adverse drug reactions in elderly hospitalized patients. Drugs Aging.
2011;28(5):379–90.
21. Violan C, Foguet-Boreu Q, Flores-Mateo G, Salisbury C, Blom J, Freitag M, et al. Prevalence,
determinants and patterns of multimorbidity in primary care: a systematic review of observa-
tional studies. PLoS One. 2014;9(7):e102149.
22. Bourgeois FT, Shannon MW, Valim C, Mandl KD. Adverse drug events in the outpatient set-
ting: an 11-year national analysis. Pharmacoepidemiol Drug Saf. 2010;19(9):901–10.
23. Corsonello A, Pedone C, Corica F, Incalzi RA. Polypharmacy in elderly patients at discharge
from the acute care hospital. Ther Clin Risk Manag. 2007;3(1):197.
24. Guthrie B, Makubate B, Hernandez-Santiago V, Dreischulte T. The rising tide of polyphar-
macy and drug–drug interactions: population database analysis 1995–2010. BMC Med.
2015;13(1):74.
25. Johnell K, Klarin I. The relationship between number of drugs and potential drug–drug inter-
actions in the elderly. Drug Saf. 2007;30(10):911–8.
26. Aapro MS, Köhne C-H, Cohen HJ, Extermann M. Never too old? Age should not be a barrier
to enrollment in cancer clinical trials. Oncologist. 2005;10(3):198–204.
27. Cruz-Jentoft AJ, Carpena-Ruiz M, Montero-Errasquín B, Sánchez-Castellano C, Sánchez-
García E. Exclusion of older adults from ongoing clinical trials about type 2 diabetes mellitus.
J Am Geriatr Soc. 2013;61(5):734–8.
80 M. Randles and D. O’Mahony
28. Rawlins M, Thompson J. Pathogenesis of adverse drug reactions. In: Davies D, editor.
Textbook of adverse drug reactions. Oxford: Oxford University Press; 1977. p. 10.
29. Aronson JK, Ferner RE. Joining the DoTS: new approach to classifying adverse drug reac-
tions. BMJ. 2003;327(7425):1222–5.
30. Coleman JJ, Pontefract SK. Adverse drug reactions. Clin Med (Lond). 2016;16(5):481–5.
31. O’Connor MN, Gallagher P, Byrne S, O’Mahony D. Adverse drug reactions in older patients
during hospitalisation: are they predictable? Age Ageing. 2012;41(6):771–6.
32. Budnitz DS, Lovegrove MC, Shehab N, Richards CL. Emergency hospitalizations for adverse
drug events in older Americans. N Engl J Med. 2011;365(21):2002–12.
33. Howard RL, Avery AJ, Slavenburg S, Royal S, Pipe G, Lucassen P, et al. Which drugs cause pre-
ventable admissions to hospital? A systematic review. Br J Clin Pharmacol. 2007;63(2):136–47.
34. Lavan AH, Gallagher P. Predicting risk of adverse drug reactions in older adults. Ther Adv
Drug Saf. 2016;7(1):11–22.
35. Naranjo CA, Busto U, Sellers EM, Sandor P, Ruiz I, Roberts EA, et al. A method for estimat-
ing the probability of adverse drug reactions. Clin Pharmacol Ther. 1981;30(2):239–45.
36. WHO. The use of the WHO–UMC system for standardised case causality assessment: World
Health Organisation. https://www.who.int/medicines/areas/quality_safety/safety_efficacy/
WHOcausality_assessment.pdf.
37. Hartwig SC, Siegel J, Schneider PJ. Preventability and severity assessment in reporting adverse
drug reactions. Am J Hosp Pharm. 1992;49(9):2229–32.
38. Hallas J, Harvald B, Gram L, Grodum E, Brøsen K, Haghfelt T, et al. Drug related hospital
admissions: the role of definitions and intensity of data collection, and the possibility of pre-
vention. J Intern Med. 1990;228(2):83–90.
39. O'Connor MN, O'Sullivan D, Gallagher PF, Eustace J, Byrne S, O'Mahony D. Prevention of
hospital-acquired adverse drug reactions in older people using screening tool of older persons’
prescriptions and screening tool to alert to right treatment criteria: a cluster randomized con-
trolled trial. J Am Geriatr Soc. 2016;64(8):1558–66.
40. Blum MR, Sallevelt BT, Spinewine A, O’Mahony D, Moutzouri E, Feller M, et al. Optimizing
therapy to prevent avoidable hospital admissions in multimorbid older adults (OPERAM):
cluster randomised controlled trial. BMJ. 2021;374:n1585.
41. Onder G, Petrovic M, Tangiisuran B, Meinardi MC, Markito-Notenboom WP, Somers A,
et al. Development and validation of a score to assess risk of adverse drug reactions among
in-hospital patients 65 years or older: the GerontoNet ADR risk score. Arch Intern Med.
2010;170(13):1142–8.
42. Tangiisuran B, Scutt G, Stevenson J, Wright J, Onder G, Petrovic M, et al. Development and
validation of a risk model for predicting adverse drug reactions in older people during hospital
stay: Brighton adverse drug reactions risk (BADRI) model. PLoS One. 2014;9(10):e111254.
43. O'Mahony D, O'Connor MN, Eustace J, Byrne S, Petrovic M, Gallagher P. The adverse
drug reaction risk in older persons (ADRROP) prediction scale: derivation and prospective
validation of an ADR risk assessment tool in older multi-morbid patients. Eur Geriatr Med.
2018;9(2):191–9.
44. Jennings E, Gallagher P, O’Mahony D. Detection and prevention of adverse drug reactions in
multi-morbid older patients. Age Ageing. 2018;48(1):10–3.
45. Killin L, Hezam A, Anderson KK, Welk B. Advanced medication reconciliation: a systematic
review of the impact on medication errors and adverse drug events associated with transitions
of care. Jt Comm J Qual Patient Saf. 2021;47(7):438–51.
Adherence: How to Measure
and Improve It 7
Alessandra Marengoni and Laura J. Sahm
A. Marengoni (*)
Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
e-mail: [email protected]
L. J. Sahm
School of Pharmacy, University College Cork, Cork, Ireland
Department of Pharmacy, Mercy University Hospital, Cork, Ireland
e-mail: [email protected]
medication, they would also not like to admit this as it may cause some embarrass-
ment or perhaps a fear of disappointing the prescriber. In addition, we know that the
more medications a patient needs, due to multimorbidity and/or older adult with
polypharmacy, the more likely that medication adherence will be suboptimal [10].
There can be the direct measurement of the amount of drug in the patient at a par-
ticular point in time, for example, by taking a blood sample. This is commonly used
when dealing with narrow therapeutic index drugs, for example, digoxin or theoph-
ylline, and there is a need to establish the therapeutic drug concentration, to avoid
sub- or supra- therapeutic doses. This can also be used to measure adherence but is
not done routinely in practice due to its cumbersome and costly nature, in addition
to the fact that parameters such as the absorption, distribution, metabolism and
elimination of the drug (or indeed its metabolites) may have individual variation.
Also, bearing in mind that the patient may have taken this drug with food or other
drugs, there may well be drug–drug or drug–food or indeed drug–herbal interac-
tions. These interactions may result in the faster/slower elimination of the drug and,
therefore, muddy the water.
At best these drug levels provide a snapshot of what is happening in the body at
the time and do not offer a view as to the long-term medication adherence. This is
analogous to the doctor who relies on a fasting blood glucose to indicate the adher-
ence of a patient to their antidiabetic agents rather than seeking to review the glyco-
sylated haemoglobin. This method may be appropriate for the measurement of
adherence to single drug therapy but does little to offer explanations as to why the
patient may not be adhering to therapy or any indication of whether this non-
adherence is a single event or indicative of someone who is persistently non-
adherent. The simplistic nature of this result, that is, plasma level of drug detected,
also fails to consider the myriad factors that go into the patient’s intentional or
unintentional non-adherence to the prescribed regimen.
I remember a specific instance when I was monitoring warfarin (vitamin K
antagonist) for a patient, and this is commonly done with the aid of an automated
decision-making tool to aid dose adjustment. The patient had a target Internationalized
Normalized Ratio (INR) of 2.5 with a range of 2–3. On this occasion, the patient
said that he was wondering whether he could consume alcohol. I answered with my
usual “warfarin loves everything but change” and that a moderate amount of alcohol
(less than two units per day) should be fine. The following week, the patient pre-
sented with an INR of 4.5. I asked whether there had been any variations in diet/
coprescribed medications/taking too many tablets and of course alcohol consump-
tion. The patient told me that he had stuck with the guidance and that he had saved
up his two units of alcohol per day and had consumed 14 units of alcohol the night
before; this was a big lesson for me and one that helps to illustrate the importance
of clear communication with our patients. Needless to say, I modified this message
going forward!
84 A. Marengoni and L. J. Sahm
These tend to be more widely used research and include the use of electronic health
records (EHR) electronic monitoring devices, pill count and self-reported measures.
From an adherence point of view, we talk about primary and secondary non-
adherence. Primary non-adherence would be the patient who visits their physician
and receives a prescription for medication but does not take this to be dispensed. In
other words, the non-adherence is clear from the first point in the sequence.
Secondary non-adherence is whereby this patient does go to the pharmacy and gets
the medication order filled, but having brought the medications home, the medica-
tion is not taken at all or not taken as directed.
If we look at the data available from pharmacy claims databases, we can estimate
what is often termed the Medication Possession Ration (MPR). The MPR has been
defined as the proportion (or percentage) of days’ supply obtained during a speci-
fied time or over a period of refill intervals [4]. Another method, the Proportion of
Days Covered (PDC) is the number of days when the medication was available
divided by the number of days in the study period. A cut-off point is commonly
advised (e.g. at least 80% adherent), and this allows the group to be divided into
adherent and non-adherent [12]. For both the MPR and the PDC, it is important to
be clear on how the calculation is performed as this can vary between studies. These
measures of adherence may be useful for seeing trends in a particular population,
but as we know just because a patient has collected the medicine, it does not mean
that it will be taken. The MPR and PDC determine the maximum adherence possi-
ble; in other words, if a patient has only collected 30 days of medication in the last
60 days, they are unable to be more than 50% adherent. Conversely, if pharmacists
were to interrogate their own data, they would be able to find individual patients
who may benefit from a medicine use review (MUR).
A MUR is a planned face-to-face consultation between a pharmacist and a patient
to discuss their medicines, both prescribed and non-prescribed. The review is medi-
cation adherence-centred and aims to help increase patients’ knowledge and under-
standing of their medicines, including how and why they should be taken. It also
7 Adherence: How to Measure and Improve It 85
review article by Nguyen et al. in which the authors have summarized the main
measures as well as their advantages and disadvantages for different cohorts [18].
Many of these questionnaires require the patient to self-complete introducing more
variability and challenges. If we consider those who are (1) non-native speakers, (2)
have low health literacy and/or specifically in the older adult, practicalities such as
being able to read the font size and/or ability to accurately recall when and how
many medications were taken in the last day(s), we can see that likelihood of having
any meaningful data is quite low. Certainly, these measures may be useful in clinical
practice by providing an opportunity to discuss any obstacles to medication adher-
ence; however, they would likely lead to inaccuracies and probable overestimation
of medication adherence due to the challenges mentioned above and social desir-
ability reporting bias. In addition, not all these questionnaires are free from restric-
tions regarding their use even for research purposes, so readers should consult the
copyright and licensing rules prior to any inclusion in a protocol.
In summary, it is likely that a multi-modal approach encompassing objective and
subjective measures will likely yield the most accurate estimate of adherence.
The benefits of proper adherence to treatment are several, including preventing dis-
eases, improvement of health, better quality of life and a more cost-effective use of
healthcare resources [19]. To date, no single intervention strategy has proven effec-
tiveness in enhancing adherence across all patients, clinical conditions and settings.
Unfortunately, a number of knowledge gaps still exist and limit previous clinical
trials aiming to improve adherence as well as adherence in the real world popula-
tion; for instance, poor understanding of the interplay between adherence, multi-
morbidity and polypharmacy, fragmentation of care and poor communication
among care players, the lack of definition of a specific target population, absence of
validated information on new technologies and lack of standardized information
systems [6].
In a Cochrane review including randomized clinical trials of interventions to
improve adherence, measuring both medication adherence and clinical outcome,
with at least 80% follow-up of each group studied and at least 6 months’ follow-up
for studies with positive findings at earlier time points, only few randomized clinical
studies showed a low risk of bias and among them only five improved both adher-
ence and clinical outcome [20]. A narrative review focused on the most frequent
interventions employed to improve medication adherence and the outcomes
achieved, with a special eye on factors associated with ageing and with cognitive
impairment or dementia [21]. The authors underlined that older persons have been
systematically excluded from randomized clinical trials especially if affected by
multimorbidity and prescribed with polypharmacy. As a consequence, the onset of
unknown adverse drug reactions during the marketing of the drugs limit adherence
in this group of patients. Regarding cognition and drug consumption, a systematic
evidence-based review showed the importance of relational aspects in improving
7 Adherence: How to Measure and Improve It 87
adherence in patients with dementia, which may benefit more from human commu-
nication as reminder than technology [22]. Indeed, in the same review, only three
studies met inclusion criteria of interventional, two evaluated reminder systems and
showed no benefit, whereas one study improved adherence through telephone and
televideo reminders at each dosing interval. Another reason of poor adherence in
older persons living in countries without welfare is the limitation to medications
access, the so-called cost-related non-adherence. The Medicare Current Beneficiary
Survey showed that in 2016, 34.5% of enrollees under 65 years with disability and
14.4% of those 65 years and older did not take their medications as prescribed due
to high costs [23]. Three indicators of worse health (general health status, functional
limits and count of conditions) were all independently associated with higher risk of
cost-related non-adherence. Strategies that effectively improve adherence could
produce financial benefits by attenuating the effect of risk factors and preventing
disease progression in a wide number of patients and different settings [24].
A few years ago, a group of professionals with expertise in geriatrics, pharmacol-
ogy, epidemiology and public health reached a consensus on the possible best inter-
ventions to improve adherence in older individuals. Seven strategies were identified
and classified based on their target (patient, therapy and public health/society)
(Table 7.2): (1) Comprehensive Geriatric Assessment (CGA)—CGA should be
administered to all older adults participating in a program aimed at improving medi-
cation adherence because it identifies their needs and priorities; (2) patient and care-
giver education—educational programs should account for health literacy level and
include lifestyle recommendations in order to improve patient empowerment; (3)
optimization of treatment—optimized treatment should be reached through clinical
decision, use of criteria for appropriate prescription and computerized prescription
support systems; (4) use of adherence aids—electronic support devices for patients,
particularly alerting non-adherence systems, web-based software tools and wear-
able units need to be implemented; (5) physicians and other healthcare profession-
als should be trained by educational professionals through a cascade model and
regular meetings focused on improving both medication knowledge and communi-
cation skills; (6) adherence assessment—adherence assessment should be mostly
objective (e.g. drug refill, clinical assays) and continuous; (7) facilitating access to
medicine by service integration by medication delivery and general practitioner–
pharmacist bidirectional consultation [6]. The authors suggested that the reasons of
non-adherence are so multifactorial that interventions to improve it should be
patient-centred, involving multiple stakeholders, with the different players provid-
ing different services, but integrated with one another. Further, in the near future,
information and communication technology (ICT) systems may become particu-
larly relevant for improving adherence, through the use of Tele-Medicine and
Internet-linked clinical support models, wireless sensor networks and so forth. ICT
technologies may provide self-management utilities for patients and caregivers;
education modules for patients, caregivers, and health professionals; and informa-
tion integration for patient care by physicians and nurses.
At present, another important issue that may limit adherence improvement at
population level is the wide variability in methodology used to evaluate adherence
Table 7.2 Interventions to improve adherence [6]
88
Health professional
Definition Target population involved How Time
Intervention 1 Comprehensive geriatric All older adults GP + healthcare assistant InterRai + tools to assess Program entry and then
Assessment (including adherence (i.e. MARS) every year or if change in
adherence behaviour) health status occurs
Intervention 2 Patient (and caregiver if Patients and caregivers or GP + healthcare assistant, Health beliefs model, Continuously
needed) education to all older and willing to pharmacist, healthcare training, multilevel
improve patient receive the intervention system, patients interventions
empowerment associations
Intervention 3 Optimization of treatment All older adults GP + healthcare assistantSoftware (i.e. Program entry and then
(reduction of CDSS) + clinical every year or if change in
polypharmacy) judgement health status occurs
Intervention 4 Use of adherence aids Older adults with Pharmacist, ICT company, Pillboxes, reminders, Continuously
cognitive impairment; pharma company blistering by the pharmacy
disability; social isolation;
polypharmacy; unintended
non-adherence
Intervention 5 Physician and other Physician and other Educational professional Cascade model; circle Continuously
healthcare professional healthcare professional and professional meetings (both focused on
education associations improved medication
knowledge and
communication skill)
Intervention 6 Adherence assessment All older adults GP + healthcare assistant; Objective (drug refill; Continuously
pharmacist drug count; clinical
assays) and subjective (i.e.
MARS/communication)
evaluation
Intervention 7 Facilitating access to All older adults GP and pharmacists ICT, medication delivery, Continuously
medicine by service GP-pharmacist
integration bidirectional consultation
A. Marengoni and L. J. Sahm
7 Adherence: How to Measure and Improve It 89
and poor reporting of findings from research on this topic. An international panel of
experts elaborated guidelines to improve the quality of drug adherence research
reporting, the ESPACOM Medication Adherence Reporting Guidelines (EMERGE)
[25], where ESPACOM is the European Society for Patient Adherence (www.espa-
com.eu). EMERGE provides guidance on reporting observational and interven-
tional studies on medication adherence useful for different stakeholders, such as
investigators, authors, journal editors and health policy decision makers.
Finally, a relevant question to be answered is whether higher adherence to medi-
cal prescription would produce better outcomes in older subjects, not only in term
of length of life, symptoms control and healthcare utilization [26], but also as ben-
eficial on functional outcomes, such as falls and cognitive decline. In a study of
community-dwelling older people presenting a prescription for antihypertensive
medication to 106 community pharmacies in Ireland, with no evidence of cognitive
impairment, taking antihypertensive medication for ≥1 year (n = 938), each 5-day
gap in antihypertensive refill adherence increased the risk of self-reported injurious
falls by 18% [27].
7.4 Conclusions
References
1. Marengoni A, Onder G. Guidelines, polypharmacy, and drug–drug interactions in patients with
multimorbidity. BMJ. 2015;350:h1059.
2. Onder G, Bonassi S, Abbatecola AM, Folino-Gallo P, Lapi F, Marchionni N, Geriatrics
Working Group of the Italian Medicines Agency, et al. High prevalence of poor-quality drug
prescribing in older individuals: a nationwide report from the Italian Medicines Agency
(AIFA). J Gerontol A Biol Sci Med Sci. 2014;69(4):430–7.
3. Onder G, Marengoni A, Russo P, Degli Esposti L, Fini M, Monaco A, Geriatrics Working
Group of the Italian Medicines Agency (Agenzia Italiana del Farmaco, AIFA), Medicines
Utilization Monitoring Center Health Database Network, et al. Advanced age and medication
prescription: more years, less medications? A nationwide report from the Italian Medicines
Agency. J Am Med Dir Assoc. 2016;17(2):168–72.
4. Growdon ME, Gan S, Yaffe K, Steinman MA. Polypharmacy among older adults with
dementia compared with those without dementia in the United States. J Am Geriatr Soc.
2021;69(9):2464–75.
5. https://apps.who.int/iris/handle/10665/42682. Accessed 14 Dec 2022.
6. Marengoni A, Monaco A, Costa E, Cherubini A, Prados-Torres A, Muth C, et al. Strategies to
improve medication adherence in older persons: consensus statement from the Senior Italia
Federanziani Advisory Board. Drugs Aging. 2016;33(9):629–37.
90 A. Marengoni and L. J. Sahm
8.1 Introduction
As older people age, the complexity of their medication use often increases as does
the frequency of healthcare utilisation. The onset of additional age-related morbidity,
destabilisation of chronic conditions and increase in frequency of hospitalisation and
healthcare utilisation can add to the older person’s medication complexity. Previous
research shows that at least 10% of hospital admissions in older people are due to
adverse drug reactions and that medication error occurs in 50% of cases following
hospital discharge [1, 2]. Therefore, strategies are needed to prevent, recognise and
manage medication-related problems prevalent in older patients at points of care tran-
sition and throughout the clinical care journey. Medication review and medication
reconciliation are processes within an overall medication management strategy that
address these issues and collectively aim to improve the older person’s transitional
medication safety (Fig. 8.1). This chapter describes these processes individually and
Medication optimisation
A series of processes to support people to get the most out of their medication
8.2.1 Theory
8.2.2 Practice
The MedRec process should ideally be applied at each point of care transition. For
example, it should be applied at hospital admission, at internal transfers and then
again at hospital discharge (Fig. 8.2). MedRec by itself is quite a transactional pro-
cess, composed of three steps: (1) collect and create, (2) check by comparing and
(3) document and communicate [5].
Step 1: Collect and Create Collect information and create the person’s Best Possible
Medication History (BPMH). This involves building a comprehensive list of the med-
ications that the person was using immediately prior to the care transition, for exam-
ple, pre-admission. This is usually created by discussion with the patient or their
representative and by obtaining information from other sources, for example, the per-
son’s community pharmacist or general practitioner, or an electronic medication
record. At least two reference sources should be consulted to build a complete list, one
of which is the patient/proxy representative. Importantly, this step should create a list
of the medication that the person was actually taking, which can differ from what was
prescribed for or dispensed to them. This may uncover potential medication non-
adherence; the reader should cross refer to Chap. 7 for information about how to
improve adherence.
Step 2: Check by Comparing This is the reconciliation step; it involves either a proac-
tive or a reactive MedRec process (Fig. 8.2). The proactive process sees the BPMH built
before the admission orders are made and the retroactive process sees the BPMH built
after the admission orders are made. In both cases, the BPMH and the post-transition
8 Medication Reconciliation and Review: Theory, Practice and Evidence 93
Fig. 8.2 An overview of medication optimisation across care transition. BPMH best possible
medication history, MO medication order
Discrepancy
Resolved
Intended
and supplement products of last or stop
dose
No preadmission medication
Unchanged
New
Changed
products
and respiratory inhalers for a patient admitted with a transient ischaemic attack.
The causes of unintentional discrepancies are multi-fold and may relate to the
patient, the healthcare professionals, the environment or the organisational
care issues.
Medication discrepancies are also categorised by the nature of the difference, the
most common being omission of medication, followed by differences in the dosage,
frequency, formulation or route of administration, and finally commission of medi-
cation, meaning the prescribing or ordering of a medication which the patient is not
actually using. Medication discrepancies are a necessary part of medication optimi-
sation and are often a clinically appropriate response to a comprehensive medica-
tion review (Fig. 8.2). They become problematic if they are errors (unintentional
discrepancies) or if intentional but not documented/communicated (Step 3).
Previous studies identified that both unintentional discrepancies and undocumented
intentional discrepancies persist across care transitions into the next care setting or
episode, potentially exposing the patient to medication-related harm due to incor-
rect administration or omission of medication.
8.2.3 Evidence
Several systematic reviews have explored the impact of undertaking the MedRec
process, few of which have focussed exclusively on older people [6–12]. Most
reviews have focussed on the hospital admission and discharge transitions and less
on MedRec following hospital discharge or transition to other care settings. Most
have investigated the benefit of MedRec as a stand-alone intervention whilst a
minority have investigated the benefit of complex transitional care interventions that
include MedRec as a component. MedRec interventions have been delivered by a
variety of healthcare personnel, most commonly pharmacy, often working interpro-
fessionally, and some studies have investigated electronic solutions.
Medication discrepancy, a process measure, is the most reported outcome in
MedRec studies. The available evidence identifies that MedRec alone may reduce
the presence of any and sum of medication discrepancies at care transition. However,
the certainty of this evidence is low because of the generally poor quality of the
available studies. The evidence of a direct benefit of MedRec on clinical outcomes,
such as adverse events or healthcare utilisation, is less clear, as is the effect on
humanistic outcomes such as quality of life. One systematic review which explored
interventions that included activities focussed on medication continuity delivered
during hospital stay or following discharge identified that MedRec reduced hospital
readmissions [12]. Interventions found to best support older patients’ medication
continuity and to have the greatest impact on reducing hospital readmission are
those that bridge transitions and include patient self-management, telephone fol-
low-up and MedRec activities.
Qualitative research studies, however, enable a broader understanding of the out-
comes affected by MedRec at care transitions, for example, the patient’s or informal
carer’s experience, activation, involvement in decision-making, workload around
managing medication and knowledge and confidence in medication use at periods
around care transitions [13].
96 T. Grimes and C. Ryan
• Medication-related
–– Increasing number of pre-admission drugs
–– Increasing number of high-risk drugsa
–– Class of medication: intake of four of the following classes, gout medication,
muscle relaxants, lipid lowering agents, antidepressants and respiratory
medication
• Patient-related
–– Age 65+ years is associated with discrepancy compared to younger people,
but amongst extremely older (>85) people, advancing age is associated with
fewer discrepancies
–– Use of multiple outpatient pharmacies
• Setting-related
–– Lack of availability of a medication list
–– Family member or caregiver consulted as an information source
As defined on the Institute for Safe Medication Practices high-alert or the North
a
8.3.1 Theory
Table 8.2 ‘Medicines review’ and international equivalent terms used defined
Organisation, country Term used Definition
Pharmaceutical care ‘Medication ‘A structured evaluation of a patient’s medicines
network Europe, review’ with the aim of optimising medicines use and
European-wide improving health outcomes. This entails detecting
drug related problems and recommending
interventions’
Pharmaceutical Society ‘Home ‘Systematic assessment of a consumer’s
of Australia’s (PSA), medicines medications and the management of those
Australia review’ medications, with the aim of optimising consumer
health outcomes and identifying potential
medication-related issues within the framework of
the quality use of medicines’
National Institute for ‘Medication ‘A structured, critical examination of a patient’s
Health and Care review’ medicines with the objective of reaching an
Excellence (NICE, UK), agreement with the patient about treatment,
United Kingdom optimising the impact of medicines, minimising
the number of medication related problems and
reducing waste’
United States ‘Medication ‘A distinct service or group of services that
therapy optimise therapeutic outcomes for individual
management’ patients’
8.3.2 Practice
medication review may warrant further intervention at the drug, patient or prescriber
level. Such intervention may trigger the need for an additional process such as the
management of an interaction (Chap. 3), an inappropriate medication (Chap. 4), a
prescribing cascade (Chap. 5), an adverse effect (Chap. 6), adherence (Chap. 7) or
deprescribing (Chap. 10), and the reader should consult these chapters for fur-
ther detail.
8.3.3 Evidence
Table 8.4 Risk patients and risk medications to target for medication review in primary care
Risk medication Risk patients
Medication increasing risk of falls Frail patients
Anticoagulant medication Patients recently discharged from hospital
Blood pressure lowering medication Patients with multimorbidity
Antidiabetic medication
Medication for Parkinson’s disease
102 T. Grimes and C. Ryan
8.4 Summary
References
1. Alqenae FA, Steinke D, Keers RN. Prevalence and nature of medication errors and medication-
related harm following discharge from hospital to community settings: a systematic review.
Drug Saf. 2020;43(6):517–37.
2. Oscanoa T, Lizaraso F, Carvajal A. Hospital admissions due to adverse drug reactions in the
elderly. A meta-analysis. Eur J Clin Pharmacol. 2017;73(6):759–70.
3. National Institute for Health and Care Excellence. Medicines optimisation: the safe and effec-
tive use of medicines to enable the best possible outcomes. NICE Guideline 5. https://www.
nice.org.uk/guidance/ng5. Accessed 27 Feb 2022.
4. Penm J, Vaillancourt R, Pouliot A. Defining and identifying concepts of medication reconcilia-
tion: an international pharmacy perspective. Res Soc Adm Pharm. 2019;15(6):632–40. https://
doi.org/10.1016/j.sapharm.2018.07.020.
5. Institute for Safe Medication Practices Canada. Medication reconciliation in acute care: getting
started kit. Canada: Institute for Safe Medication Practices; 2011. https://www.ismp-canada.
org/download/MedRec/Medrec_AC_English_GSK_V3.pdf. Accessed 27 Feb 2022
6. Anderson LJ, Schnipper JL, Nuckols TK, Shane R, Le MM, Robbins K, et al. Effect of medi-
cation reconciliation interventions on outcomes: a systematic overview of systematic reviews.
Am J Health Syst Pharm. 2019;76(24):2028–40. https://doi.org/10.1093/ajhp/zxz236.
7. Guisado-Gil AB, Mejías-Trueba M, Alfaro-Lara ER, Sánchez-Hidalgo M, Ramírez-Duque
N, Santos-Rubio MD. Impact of medication reconciliation on health outcomes: an overview
of systematic reviews. Res Soc Adm Pharm. 2020;16(8):995–1002. https://doi.org/10.1016/j.
sapharm.2019.10.011.
8. Killin L, Hezam A, Anderson KK, Welk B. Advanced medication reconciliation: a sys-
tematic review of the impact on medication errors and adverse drug events associated with
8 Medication Reconciliation and Review: Theory, Practice and Evidence 103
26. Abrahamsen B, Hansen RN, Rossing C. For which patient subgroups are there posi-
tive outcomes from a medication review? A systematic review. Pharm Pract (Granada).
2020;18(4):1976. https://doi.org/10.18549/pharmpract.2020.4.1976.
27. Kempen TG, Hedström M, Olsson H, Johansson A, Ottosson S, Al-Sammak Y, et al.
Assessment tool for hospital admissions related to medications: development and validation in
older patients. Int J Clin Pharm. 2019;41(1):198–206.
28. Thevelin S, Spinewine A, Beuscart JB, Boland B, Marien S, Vaillant F, et al. Development of a
standardized chart review method to identify drug-related hospital admissions in older people.
Br J Clin Pharmacol. 2018;84(11):2600–14.
The Role of Pharmacists in Optimising
Drug Therapy 9
Anne Spinewine, Stephen Byrne, and Olivia Dalleur
A. Spinewine · O. Dalleur
Université catholique de Louvain, Louvain Drug Research Institute and Faculty of Pharmacy
and Biomedical Sciences, Brussels, Belgium
e-mail: [email protected]; [email protected];
[email protected]; [email protected]
S. Byrne (*)
Pharmaceutical Care Research Group, School of Pharmacy, University College Cork,
Cork, Ireland
e-mail: [email protected]
HCP at the time of the review. The Pharmaceutical Care Network European (PCNE)
group in 2018 also agreed through their consensus process on four subtypes of
medication reviews, ranging from those conducted in a community pharmacy, to
more comprehensive reviews, which require full access to patient’s medical record
(e.g. access to medical notes in hospital, long-term care or general practice set-
tings). The subclassification of a medication review is as follows:
• “PCNE Type 1: Simple MR: A simple medication review is based on the avail-
able medication history in the pharmacy; this type of review occurs routinely
during the medication supply/dispensing process, for example, drug–drug inter-
actions, reviewing the appropriateness of a particular medication dosages, poten-
tial side effects and medication adherence issues.”
• “PCNE Type 2A: Intermediate MR: An intermediate medication review can be
performed when the patient can be approached for information. Such a review is
based on medication history and patient information.” Which builds upon a Type
1 review and expands the review to include issues such as drug–food interac-
tions, issues with over-the-counter (OTC) medication and medication effective-
ness issues.
• “PCNE Type 2B: Intermediate MR: An intermediate medication review can be
performed if information from the general practitioner is also available. Such a
review is based on medication history and medical information.” This type of
review builds upon Type 2A by including the identification of potential medi-
cines without a current clinical indication.
• “PCNE Type 3: Advanced MR: An advanced medication review is based on
medication history, patient information and clinical information. This type of
review is the most comprehensive and in-depth review and results in all of the
above being addressed with the addition of the identification of an indication
without a potential medication being prescribed for its treatment [1].
ensuring the patient has been educated about the discharge treatment plan; (f) dis-
pensing an adequate amount of medication at discharge and (g) communicating to
follow-up healthcare [5]. A core concept that is closely related to these principles is
medication reconciliation (MedRec) (see Chap. 8). MedRec has been defined as
“the process of identifying the most accurate list of all medications a patient is
taking—including name, dosage, frequency, and route—and using this list to pro-
vide correct medications for patients anywhere within the health care system” [6].
MedRec is a safety strategy adopted by most guidelines and health organisations
that should be performed at each transition, ideally within 24 h.
MedRec prevents transcribing errors, improves monitoring of prescriptions, facili-
tates switch from medication taken at home towards formulary medicines upon
admission [7]. By improving communication between HCPs and with the patient,
MedRec reduces medication discrepancies and potentially harmful medication errors.
In older patients specifically, pharmacist-led MedRec reduces medication errors [8].
However, evidence on patient-related clinical outcomes is inconclusive. Nevertheless,
a meta-analysis focusing on the effectiveness of pharmacist-led MedRec on patient-
related clinical outcomes during the transition to and from hospital settings showed a
reduction in drug-related hospital revisits, emergency department visits and in hospi-
tal readmissions [9]. Additionally, there is evidence that pharmacist-led MedRec at
discharge could be a cost-effective intervention by reducing readmissions [10].
Many pharmaceutical societies provide guidelines, practical tools and educa-
tional support for MedRec implementation. Strategies for implementing MedRec
include the use of standardised forms, electronic reconciliation tools, collaborative
models and patient engagement.
After discharge, many patients might have difficulties with medication manage-
ment, including delays in filling their medications. Therefore, at discharge, addi-
tional pharmacists’ interventions complement MedRec to improve continuity of
care, often evaluated in the literature in combination: supplying of medications,
medication counselling of patients, provision of verbal and written information to
the patient, communication between primary and secondary care providers, follow-
up phone calls with the patient or the community pharmacist and home visits. These
interventions are particularly recommended in older patients and can be performed
by both the hospital pharmacist and the community pharmacist.
The written report that all patients and subsequent HCPs receive at discharge, also
called discharge plan, contains a detailed list of all medications the patient is currently
taking (name and respective indications, strength, form, timing, frequency, duration,
date and time of the last dose), identification and explanation about medicines that
have been started/stopped, any known adverse reactions and known allergies. This
report is in a format suitable and adapted to the patient’s and/or carer’s health literacy.
Medication adherence has been defined as “the process by which patients take their
medications as prescribed” [11]. Several factors influence adherence such as
108 A. Spinewine et al.
knowledge and skills in managing medications, previous side effects, health liter-
acy, beliefs about the medication and lack of involvement in the treatment decision-
making process.
Patient education is a common intervention to improve medication adherence.
Pharmacists-led education can be provided in several settings and formats: meet-
ing during hospital stay, consultations, including in community pharmacies, provi-
sion of written information, home visits, phone calls and motivational interviewing
[12]. Rather than just presenting information about medications, pharmacist dis-
cusses it and assesses non-adherence and beliefs about medication. Education to
tackle non-adherence should include tailored practical suggestion (e.g. to the type
of medicines or regimen, monitoring, use of a pill-box or reminders). Pharmacists
also implement patient self-management plans, to support people with chronic
conditions who want to be involved in managing of their own medicines. Patient
education can be general or specifically focused on oncology medications, antico-
agulation, antihypertensive drugs, analgesics, potentially inappropriate medica-
tions, fall-risk medications and so forth. Health literacy tools can help pharmacists
adapt education and counselling.
Although evidence of pharmacists-led education in older patients on adherence
and clinical outcomes is inconsistent, some studies have shown promising results on
optimisation of medication in terms of deprescribing of inappropriate medications
and benzodiazepines in ambulatory and hospital setting [13, 14].
Most patients, including the older ones, wish to be involved in making decisions
about their own medicines. Shared decision-making (SDM) is “an approach where
healthcare professionals and patients share the best available evidence when faced
with making decisions regarding healthcare, and where patients are supported to
consider options to achieve informed preferences” [15]. In older patients, SDM and
patient involvement are important to reduce inappropriate polypharmacy. Moreover,
SDM results in older patient to choose more conservative options (medication
reduction or cessation, less initiation of new drugs), which support deprescribing
and decrease of treatment burden [16, 17]. Pharmacists should integrate shared
decision-making (SDM) with the patient in their medication review and treatment
optimisation process. They can also help develop medication decision tools, which
will support discussion but not replace it.
Pharmacy teams internationally have played a pivotal role in the education and
training of fellow HCPs in both the acute and non-acute settings over several
decades. Such education and training sessions take place both in formal, for exam-
ple, lecture, and/or informal, for example, ward rounds settings, but one thing is
clear, if our desire is to improve prescribing amongst older patients, then constant
and up-to-date education of all prescribers including non-medical prescribers (e.g.
nurse and other HCP prescribers) is crucial. There have been numerous educational
9 The Role of Pharmacists in Optimising Drug Therapy 109
Clinical pharmacy services with a focus on older people (also called “geriatric phar-
macy practice”) emerged in the mid-1970s, and since then a substantial body of
published literature has accumulated, demonstrating patient safety and economic
benefits of clinical pharmacy services for older people, including evidence from
randomised controlled trials [26]. Clinical pharmacy services have been shown to
improve prescribing, prevent adverse drug reactions and medication errors, improve
patients’ medication knowledge and adherence and, in some populations, reduce the
risk of medication-related hospital admissions [26].
This movement of pharmacists’ involvement in direct patient care away from the
traditional medication-dispensing/supply functions is widespread globally, although
110 A. Spinewine et al.
there is major inter- and intra-country variability in the practice models and their
level of implementation [27, 28]. Below we describe and discuss these models of
practice in each type of setting of care.
identifies and refers the patient for MR; an accredited pharmacist visits the patient
at home and obtains a comprehensive medication history, then documents its medi-
cation review findings and recommendations in a report for the general practitioner
(GP); then based on this, the GP and patient formulate a medication plan [4]. The
positive impact has been demonstrated on medication use measures mainly [4].
Drug-related admissions, adverse drug events during hospital stay and after dis-
charge are all frequent in older people with multimorbidity and polypharmacy. The
involvement of clinical pharmacists to optimise drug therapy in older inpatients is,
therefore, appealing. Various practices for pharmacists’ involvement in the care of
inpatients exist, ranging from single approaches such as simple or intermediate MR
during hospital stay to more comprehensive, multifaceted and collaborative
approaches implemented from admission to discharge or even post-discharge.
Current evidence to date shows that medication reconciliation, patient education,
HCP education and transitional care are essential elements that need to be imple-
mented in addition to medication review to reach an effect on hospital readmissions
[33–35]. Even though there has been some progress over the last decade, such com-
prehensive model of care is not yet widely implemented in hospitals in Europe and
beyond. The role of the “ward-based clinical pharmacist,” whilst well established in
some countries, is still a rarity in others. Lack of capacity, capability and support
from managers are the commonly cited reasons for this [36, 37].
In the case of limited resources, pharmacists and hospital managers may decide
to have a comprehensive model of practice implemented for a restricted number of
older people at highest risk of drug-related problems, or to have a less comprehen-
sive and more diluted approach implemented for a larger population. The former
approach is probably preferable, although there are no strong direct comparison
data available to date to confirm this.
Polypharmacy, together with other factors such as frailty and complexity of the
medication use process, makes the safe use of medications for nursing home resi-
dents highly challenging, and pharmacists have been involved in many initiatives
aiming to optimise the use of medicines in that setting. Pharmacists’ contribution to
optimise medicines use in the nursing home setting includes (a) practices directed
at nursing home residents, often in the form of medication reviews, which can be
uni- or multiprofessional, and aimed at specific drugs or the whole regimen and (b)
practices directed at HCPs—often educational approaches directed at prescribers
[19, 38]. These practices are described further below. Even though most activities
performed to date by pharmacists focus on the prescription component, there is
wide room for pharmacists’ input towards improving administration
112 A. Spinewine et al.
process—although only limited data are available on the practice models and evi-
dence of effect [38].
In several countries such as the United Kingdom, Australia and the United
States, comprehensive MR programs in nursing homes are funded. MRs are gener-
ally conducted collaboratively by the resident’s general practitioner and a pharma-
cist who is accredited to conduct MRs (also referred to as consultant pharmacists).
As an example, Australian guidelines recommend that a MR (called Residential
Medication Management Review—similar to the Home Medicines Review pro-
gram funded in ambulatory care) is provided as soon as possible after an individual
first enters permanent residential care and when clinical circumstances change [4,
39]. Each review identifies an average of 2.7–3.9 medication-related problems per
resident receiving the service, with general practitioners accepting 45–84% of
pharmacists’ recommendations [40]. Targeted MRs for people taking psychotropic
drugs is also recommended and encouraged in several countries. There is evidence
that such programs lead to more appropriate prescriptions and lower use of seda-
tive and anticholinergics drug use, although certainty of the evidence was found to
be low in a Cochrane review [19]. Some recent clinical trials showed positive
results of collaborative and innovative approaches towards MR in the nursing
home setting, for example, by involving nursing home residents and/or nurses in
the process [41, 42]. Wider scale implementation of these approaches has yet
to occur.
Pharmacists also contribute to optimised medication use in nursing homes
through educational approaches, case conferencing, audit and feedback directed at
other HCPs (or sometimes at nursing home residents and their families) [18, 28].
Such practices are less resource intensive—and may, therefore, be encouraged in
countries where there is no government-funded MR program. They may contribute
to better use of medicines, although probably with lower effect than comprehensive
MR. As an example, in Switzerland there is funding for regular meetings (also
called quality circles) between pharmacists, physicians and nurses active in a nurs-
ing home, with the goal of producing local prescribing consensus to improve drug
use [43]. This approach led, in particular, to a reduction in drug costs and improved
antibiotics stewardship.
Pharmacists have distinct expertise that can contribute to team knowledge and com-
petence in managing older patients with polypharmacy and multiple chronic condi-
tions. Importantly, evidence shows that globally, interventions developed by a
pharmacist within a multidisciplinary team seem to provide greater benefits in terms
of clinical outcomes and economic results than those carried out by individual phar-
macists [34].
Pharmacists can be members of geriatric evaluation and management (GEM)
teams. Such teams provide comprehensive geriatric assessment, which is by nature
9 The Role of Pharmacists in Optimising Drug Therapy 113
Traditionally the prescribing role has being a key domain of the medical practitio-
ner, although globally numerous healthcare systems have broadened this authority
to other HCPs, for example, nurses, pharmacist, physiotherapist, in the form of
supplementary or independent prescribing [47]. The expansion of such authority
has demonstrated several benefits in terms of increased patient satisfaction, cost
effectiveness [37], concordance with therapeutic regimens [48], increasing continu-
ity of care and increased medication access [49, 50]. This broadening of roles for
other HCPs has been in response to changing service needs and increased specialist
roles within these professionals [47].
Key drivers resulting in the expansion of prescription authority have been a
desire by medical providers to reduce costs whilst simultaneously enhancing patient
services, whereas a key barrier for many healthcare professions to take on such roles
in the past has been the lack of access to patient’s clinical data and medical notes.
Without access to such data, it is hard for any professionals to provide clinically
appropriate advice pertaining to either prescribing or indeed appropriate advice in
relation to deprescribing [25, 47].
The impact of non-medical prescribing upon professional boundaries and rela-
tionships is often dependent upon whether supplementary or independent prescrib-
ing is legislated. In the former, continued medical authority/responsibility is retained
by the physician with supplementary prescribing authority given to the pharmacist
by the doctor after they have undertaken the clinical assessment and made the initial
114 A. Spinewine et al.
Pharmacists have specific knowledge and skills that are essential to optimising med-
ication use in older people. At the patient individual level, pharmacists’ activities
most frequently encompass medication review, medication reconciliation and
patient education. Several models of care have been implemented in different set-
tings of care (ambulatory, acute and long-term care) in many countries, although
there remains a large variability in the existence and implementation for such ser-
vices worldwide. More comprehensive, patient-centred and interprofessional
approaches seem to provide greater benefit, although there are more challenges to
implementing these approaches, especially in the ambulatory setting. Standardisation
and enhanced coverage/implementation of geriatric pharmacist practice are impor-
tant challenges for the future.
Pharmacists also make an important contribution to optimising medication use in
older people by educating other HCPs. This can happen through different approaches,
from traditional lecture sessions to more innovative approaches such as massive
open online course and academic detailing.
Finally, and even though this was not specifically addressed in the current chap-
ter, the role of pharmacists as researchers—to measure the quality of medication
9 The Role of Pharmacists in Optimising Drug Therapy 115
use, understand the barriers and enablers, and design and evaluate the effect of opti-
misation approaches—is also an important contribution towards optimising phar-
macotherapy in older patients.
References
1. Griese-Mammen N, Hersberger KE, Messerli M, Leikola S, Horvat N, van Mil JWF, et al. PCNE
definition of medication review: reaching agreement. Int J Clin Pharm. 2018;40(5):1199–208.
2. Blenkinsopp A, Bond C, Raynor DK. Medication reviews. Br J Clin Pharmacol.
2012;74(4):573–80.
3. O'Mahony D, Gudmundsson A, Soiza RL, Petrovic M, Jose Cruz-Jentoft A, Cherubini A,
et al. Prevention of adverse drug reactions in hospitalized older patients with multi-morbidity
and polypharmacy: the SENATOR* randomized controlled clinical trial. Age Ageing.
2020;49(4):605–14.
4. Chen TF. Pharmacist-led home medicines review and residential medication management
review: the Australian model. Drugs Aging. 2016;33(3):199–204.
5. Australian Pharmaceutical Advisory Council. National guidelines to achieve the continuum
of quality use of medicines between hospital and community. Canberra: Commonwealth
Department of Health and Family Services; 1998.
6. review I-IfHIMr. http://www.ihi.org:80/resources/Pages/Tools/
MedicationReconciliationReview.aspx.
7. Redmond P, Grimes TC, McDonnell R, Boland F, Hughes C, Fahey T. Impact of medication rec-
onciliation for improving transitions of care. Cochrane Database Syst Rev. 2018;8:CD010791.
8. Cameli D, Francis M, Francois VE, Medder NR, Von Eden L, Truglio-Londrigan M. A system-
atic review of medication reconciliation strategies to reduce medication errors in community
dwelling older adults. JBI Libr Syst Rev. 2012;10(42 Suppl):1–18.
9. Mekonnen AB, McLachlan AJ, Brien JA. Effectiveness of pharmacist-led medication recon-
ciliation programmes on clinical outcomes at hospital transitions: a systematic review and
meta-analysis. BMJ Open. 2016;6(2):e010003.
10. Najafzadeh M, Schnipper JL, Shrank WH, Kymes S, Brennan TA, Choudhry NK. Economic
value of pharmacist-led medication reconciliation for reducing medication errors after hospital
discharge. Am J Manag Care. 2016;22(10):654–61.
11. Vrijens B, De Geest S, Hughes DA, Przemyslaw K, Demonceau J, Ruppar T, et al. A new
taxonomy for describing and defining adherence to medications. Br J Clin Pharmacol.
2012;73(5):691–705.
12. Kini V, Ho PM. Interventions to improve medication adherence: a review.
JAMA. 2018;320(23):2461–73.
13. Marcum ZA, Hanlon JT, Murray MD. Improving medication adherence and health outcomes
in older adults: an evidence-based review of randomized controlled trials. Drugs Aging.
2017;34(3):191–201.
14. Martin P, Tamblyn R, Benedetti A, Ahmed S, Tannenbaum C. Effect of a pharmacist-led
educational intervention on inappropriate medication prescriptions in older adults: the
D-PRESCRIBE randomized clinical trial. JAMA. 2018;320(18):1889–98.
15. Joseph-Williams N, Lloyd A, Edwards A, Stobbart L, Tomson D, Macphail S, et al.
Implementing shared decision making in the NHS: lessons from the MAGIC programme.
BMJ. 2017;357:j1744.
16. Jansen J, Naganathan V, Carter SM, McLachlan AJ, Nickel B, Irwig L, et al. Too much medi-
cine in older people? Deprescribing through shared decision making. BMJ. 2016;353:i2893.
17. Tinetti ME, Naik AD, Dindo L, Costello DM, Esterson J, Geda M, et al. Association of patient
priorities-aligned decision-making with patient outcomes and ambulatory health care burden
among older adults with multiple chronic conditions: a nonrandomized clinical trial. JAMA
Intern Med. 2019;179(12):1688–97.
116 A. Spinewine et al.
18. Loganathan M, Singh S, Franklin BD, Bottle A, Majeed A. Interventions to optimise prescrib-
ing in care homes: systematic review. Age Ageing. 2011;40(2):150–62.
19. Alldred DP, Kennedy MC, Hughes C, Chen TF, Miller P. Interventions to optimise prescribing
for older people in care homes. Cochrane Database Syst Rev. 2016;2:CD009095.
20. Duckett A, Cuoco T, Pride P, Wiley K, Iverson PJ, Marsden J, et al. Academic detailing to
teach aging and geriatrics. Gerontol Geriatr Educ. 2015;36(4):331–42.
21. Lutters M, Harbarth S, Janssens JP, Freudiger H, Herrmann F, Michel JP, et al. Effect of a
comprehensive, multidisciplinary, educational program on the use of antibiotics in a geriatric
university hospital. J Am Geriatr Soc. 2004;52(1):112–6.
22. Nicolas C, Balardy L, Antoine V, Albarede J, Azria D, Cristol L, et al. Spreading geriatric
oncology culture through professional caregivers: results of a French massive open online
course (MOOC). J Geriatr Oncol. 2021;13(1):94–9.
23. O'Brien MA, Rogers S, Jamtvedt G, Oxman AD, Odgaard-Jensen J, Kristoffersen DT, et al.
Educational outreach visits: effects on professional practice and health care outcomes.
Cochrane Database Syst Rev. 2007;4:CD000409.
24. Cullinan S, O'Mahony D, Byrne S. Use of an e-learning educational module to better
equip doctors to prescribe for older patients: a randomised controlled trial. Drugs Aging.
2017;34(5):367–74.
25. Rankin A, Cadogan CA, Patterson SM, Kerse N, Cardwell CR, Bradley MC, et al. Interventions
to improve the appropriate use of polypharmacy for older people. Cochrane Database Syst
Rev. 2018;9:CD008165.
26. Elliott RA. Geriatric medicine and pharmacy practice: a historical perspective. J Pharm Pract
Res. 2016;46:9.
27. Lee JK, Slack MK, Martin J, Ehrman C, Chisholm-Burns M. Geriatric patient care by
U.S. pharmacists in healthcare teams: systematic review and meta-analyses. J Am Geriatr Soc.
2013;61(7):1119–27.
28. Spinewine A, Fialova D, Byrne S. The role of the pharmacist in optimizing pharmacotherapy
in older people. Drugs Aging. 2012;29(6):495–510.
29. Imfeld-Isenegger TL, Soares IB, Makovec UN, Horvat N, Kos M, van Mil F, et al. Community
pharmacist-led medication review procedures across Europe: characterization, implementa-
tion and remuneration. Res Soc Adm Pharm. 2020;16(8):1057–66.
30. Soares IB, Imfeld-Isenegger TL, Makovec UN, Horvat N, Kos M, Arnet I, et al. A survey to
assess the availability, implementation rate and remuneration of pharmacist-led cognitive ser-
vices throughout Europe. Res Soc Adm Pharm. 2020;16(1):41–7.
31. Latif A. Community pharmacy medicines use review: current challenges. Integr Pharm Res
Pract. 2017;7:83–92.
32. Alshehri AA, Cheema E, Yahyouche A, Haque MS, Jalal Z. Evaluating the role and integra-
tion of general practice pharmacists in England: a cross-sectional study. Int J Clin Pharm.
2021;43(6):1609–18.
33. Dautzenberg L, Bretagne L, Koek HL, Tsokani S, Zevgiti S, Rodondi N, et al. Medication
review interventions to reduce hospital readmissions in older people. J Am Geriatr Soc.
2021;69(6):1646–58.
34. Delgado-Silveira E, Velez-Diaz-Pallares M, Munoz-Garcia M, Correa-Perez A, Alvarez-Diaz
AM, Cruz-Jentoft AJ. Effects of hospital pharmacist interventions on health outcomes in older
polymedicated inpatients: a scoping review. Eur Geriatr Med. 2021;12(3):509–44.
35. Van der Linden L, Hias J, Walgraeve K, Flamaing J, Tournoy J, Spriet I. Clinical pharmacy
services in older inpatients: an evidence-based review. Drugs Aging. 2020;37(3):161–74.
36. European Association of Hospital Pharmacists aKU, Center for Medicines Optimisation.
European Statements of Hospital Pharmacy survey results 2018: statements sections 1, 3, 4.
Brussels: EAHP; 2018–2019.
37. Dalton K, Byrne S. Role of the pharmacist in reducing healthcare costs: current insights. Integr
Pharm Res Pract. 2017;6:37–46.
38. Spinewine A, Evrard P, Hughes C. Interventions to optimize medication use in nursing homes:
a narrative review. Eur Geriatr Med. 2021;12(3):551–67.
9 The Role of Pharmacists in Optimising Drug Therapy 117
39. Sluggett JK, Bell JS, Lang C, Corlis M, Whitehead C, Wesselingh SL, et al. Variation in provi-
sion of collaborative medication reviews on entry to long-term care facilities. J Am Med Dir
Assoc. 2021;22(1):148–55 e1.
40. Chen EYH, Wang KN, Sluggett JK, Ilomaki J, Hilmer SN, Corlis M, et al. Process, impact
and outcomes of medication review in Australian residential aged care facilities: a systematic
review. Australas J Ageing. 2019;38(Suppl 2):9–25.
41. Strauven G, Anrys P, Vandael E, Henrard S, De Lepeleire J, Spinewine A, et al. Cluster-
controlled trial of an intervention to improve prescribing in nursing homes study. J Am Med
Dir Assoc. 2019;20(11):1404–11.
42. Wouters H, Scheper J, Koning H, Brouwer C, Twisk JW, van der Meer H, et al. Discontinuing
inappropriate medication use in nursing home residents: a cluster randomized controlled trial.
Ann Intern Med. 2017;167(9):609–17.
43. Cateau D, Ballabeni P, Niquille A. Effects of an interprofessional quality circle-deprescribing
module (QC-DeMo) in Swiss nursing homes: a randomised controlled trial. BMC Geriatr.
2021;21(1):289.
44. Bollen A, Harrison R, Aslani P, van Haastregt JCM. Factors influencing interprofessional col-
laboration between community pharmacists and general practitioners—a systematic review.
Health Soc Care Community. 2019;27(4):e189–212.
45. Rawlinson C, Carron T, Cohidon C, Arditi C, Hong QN, Pluye P, et al. An overview of reviews
on interprofessional collaboration in primary care: barriers and facilitators. Int J Integr Care.
2021;21(2):32.
46. Sangaleti C, Schveitzer MC, Peduzzi M, Zoboli E, Soares CB. Experiences and shared mean-
ing of teamwork and interprofessional collaboration among health care professionals in pri-
mary health care settings: a systematic review. JBI Database System Rev Implement Rep.
2017;15(11):2723–88.
47. Weeks G, George J, Maclure K, Stewart D. Non-medical prescribing versus medical pre-
scribing for acute and chronic disease management in primary and secondary care. Cochrane
Database Syst Rev. 2016;11:CD011227.
48. Berry D, Bradlow A, Courtenay M. Patients’ attitudes towards, and information needs in rela-
tion to, nurse prescribing in rheumatology. J Clin Nurs. 2008;17(2):266–73.
49. Cooper R, Anderson C, Avery T, Bissell P, Guillaume L, Hutchinson A, et al. Stakeholders’
views of UK nurse and pharmacist supplementary prescribing. J Health Serv Res Policy.
2008;13(4):215–21.
50. Emmerton L, Marriott J, Bessell T, Nissen L, Dean L. Pharmacists and prescribing rights:
review of international developments. J Pharm Pharm Sci. 2005;8(2):217–25.
51. Cooper RJ, Bissell P, Ward P, Murphy E, Anderson C, Avery T, et al. Further challenges to
medical dominance? The case of nurse and pharmacist supplementary prescribing. Health
(London). 2012;16(2):115–33.
52. Iqbal MZ, Khan AH, Iqbal MS, Syed Sulaiman SA. A review of pharmacist-led interventions
on diabetes outcomes: an observational analysis to explore diabetes care opportunities for
pharmacists. J Pharm Bioallied Sci. 2019;11(4):299–309.
53. Kroezen M, Francke AL, Groenewegen PP, van Dijk L. Nurse prescribing of medicines in
Western European and Anglo-Saxon countries: a survey on forces, conditions and jurisdic-
tional control. Int J Nurs Stud. 2012;49(8):1002–12.
54. Jebara T, Cunningham S, MacLure K, Awaisu A, Pallivalapila A, Stewart D. Stakeholders’
views and experiences of pharmacist prescribing: a systematic review. Br J Clin Pharmacol.
2018;84(9):1883–905.
Deprescribing: Evidence Base
and Implementation 10
Denis Curtin and Denis O’Mahony
10.1 Introduction
Older people in their final years are commonly prescribed multiple medications to
manage their chronic diseases. These medications may ameliorate symptoms, pre-
vent future adverse health events and extend life. However, the use of multiple med-
ications is also associated with an increased risk of adverse side effects, drug
interactions and medication adherence problems. Furthermore, as older people
become increasingly frail, the use of multiple medications may be considered bur-
densome or even futile. For frailer older patients taking multiple medications, when
does the balance shift from net benefit to net harm? If declining health and death are
unavoidable, it follows logically that there must come a point in time when patients
no longer benefit from certain chronic disease drug therapies.
Chapter 4 discussed the process of deprescribing in the context of potentially
inappropriate medication (PIM) use where the goal is to minimize the adverse con-
sequences of inappropriate prescribing that is, adverse drug reactions (ADRs) and
adverse drug events (ADEs). The present chapter will focus on older people with
more advanced degrees of frailty and the discontinuation of medications that may,
ordinarily, be therapeutically useful and appropriate but of less clinical value or pos-
sibly futile in the patient who is likely to be approaching end-of-life. In this context,
the key question for the clinician is how to separate essential medications from
those that are non-essential.
The term ‘deprescribing’ is a relatively new term and first appeared in the English
language literature in 2003 [1]. In the first review of the subject, Woodward outlined
the principals of deprescribing. These included:
Since then, several new definitions have been proposed [2–4]. A 2015 system-
atic review of the literature by Reeve et al. was conducted to determine whether
a standardized definition of deprescribing could be reached to inform future
research on the subject [5]. The most common characteristics of the various defi-
nitions were used by the authors to develop a new definition: ‘Deprescribing is
the process of withdrawal of an inappropriate medication, supervised by a health
care professional with the goal of managing polypharmacy and improving
outcomes’.
from those with a low probability of dying (the clinician would be surprised, i.e.
surprise question negative [SQ−]). Recent systematic reviews of the ‘surprise ques-
tion’ indicate that while it leads to the detection of many false positives, the method
appears to be very effective at excluding patients with longer survival times (nega-
tive predictive value >90% in both reviews) [15, 16]. Clinical judgement may also
be improved by considering the illness and disability trajectory of the patient. Most
older people experience loss of functional capabilities in the last year of life and
very often this decline coincides with acute illness requiring hospitalization [15,
16]. One large Scottish study indicates that more than one in four patients aged
≥65 years admitted to hospital will be deceased in the prospective 12 months [17].
An acute hospital admission, therefore, is often a sentinel event for an older person,
especially if associated with new or additional disability, and may be an appropriate
trigger for discussions around goals of care medication rationalization.
Table 10.1 Definitions of number needed to treat (NNT), time to benefit (TTB) and time to
harm (TTH)
Definition
Number needed The number of patients that need to be treated for one patient to benefit
to treat (NNT)
Time to benefit The time taken for a statistically significant benefit to be observed in trials
(TTB) of people receiving a particular medication or therapy compared to the
control group
Time to harm The time period until a statistically significant adverse effect is observed in
(TTH) a trial within the treatment group compared to the control group
10 Deprescribing: Evidence Base and Implementation 123
primarily on the two most widely used deprescribing tools that have been tested as
interventions in well-designed clinical trials, that is, Scott’s deprescribing algorithm
and STOPPFrail criteria.
The algorithm proposed by Scott et al. (Fig. 10.1) requires the user to answer a
series of questions about each individual medication in the patient’s regimen [21].
Although comprehensive and patient-centred, the outcome is likely to depend on
the knowledge, experience and attitudes of the user. Nuanced clinical judgement is
STOPPFrail is a list of potentially inappropriate prescribing indicators designed to assist physicians with deprescribing decisions.
It is intended for older people with limited life expectancy for whom the goal of care is to optimize quality of life and minimize the risk
of drug-related morbidity. Goals of care should be clearly defined and, where possible, medication changes should be discussed and
agreed with patient and/or family. Appropriate candidates for STOPPFrail-guided deprescribing typically meet ALL of the
following criteria:
1. ADL dependency (i.e. assistance with dressing, washing, transferring, walking) and/or severe chronic disease and/or terminal illness
2. Severe irreversible frailty i.e. high risk of acute medical complications and clinical deterioration
3. Physician overseeing care of patient would not be surprised if the patient died in the next 12 months
Section A: i. Any drug that the patient persistently fails to take or tolerate despite adequate education and consideration of all
General appropriate formulations.
ii. Any drug without a clear clinical indication
iii. Any drug for symptoms which have now resolved (e.g. pain, nausea, vertigo, pruritis)
Section B: i. Lipid lowering therapies (statins, ezetimibe, bile acid sequestrants, fibrates, nicotinic acid, lomitapide, andacipimox)
Cardiology ii. Antihypertensive therapies: Carefully reduce or discontinue these drugs in patients with systolic blood pressure (SBP)
system persistently <130mmHg. An appropriate SBP target in frail older people is 130 -160mmHg. Before stopping, consider
whether the drug is treating additional conditions (e.g. beta-blocker for rate control in atrial fibrillation, diuretics for
ymptomatic heart failure)
iii. Anti-anginal therapy (specifically: nitrates, nicorandil, ranolazine): None of these anti-anginal drugs havebeen
proven to reduce cardiovascular mortality or the rate of myocardial infarction. Aim to carefully reduce and discontinue
these drugs in patients who have had no reported anginal symptoms in the previous 12 months AND who have no
proven or objective evidence of coronary artery disease.
Section C: i. Anti-platelets: No evidence of benefit forprimary (as distinct from secondary) cardiovascular prevention.
Coagulation ii. Aspirin for stroke prevention in atrial fibrillation: Aspirin has little or no role for stroke prevention in frail older
system people who are not candidates for anticoagulation therapy and may significantly increase bleeding risk.
Section D: i. Neuroleptic antipsychotics in patients with dementia: Aim to reduce dose and discontinue these drugs in patients taking them
Central for longer than 12 weeks if there are no current clinical features of behavioural and psychiatric symptoms of dementia (BPSD).
Nervous ii. Memantine: Discontinue and monitor in patients with moderate to severe dementia, unless memantine has clearly
System improved BPSD
Section E: i. Proton Pump Inhibitors: Reduce dose of Proton Pump Inhibitors when used at full therapeutic dose ≥ 8 weeks, unless
Gastrointestinal persistent dyspeptic symptoms at lower maintenance dose.
System
ii. H2 receptor antagonist: Reduce dose of H2 receptor antagonists when used at full therapeutic dose for ≥
8 weeks, unless persistent dyspeptic symptoms at lower maintenance dose.
Section F: i. Theophylline and aminophylline: These drugs have a narrow therapeutic index,have doubtful therapeutic benefit andrequire
Respiratory monitoring of serum levels and interact with other commonly prescribed drugs putting patients at an increased risk of ADEs.
System ii. Leukotriene antagonists (Montelukast, Zafirlukast): These drugs have no proven role in COPD, they are indicated only in
asthma.
Section G: i. Calcium supplements: Unlikely to be of any benefitin short-termunless proven, symptomatic hypocalcaemia.
Musculoskeletal ii. Vitamin D (ergocalciferol and colecalciferol): Lack of clearevidence to support the use of vitamin D to prevent falls and
System fractures, cardiovascular events, or cancer.
iii. Anti-resorptive/bone anabolic drugsFOR OSTEOPOROSIS (bisphosphonates, strontium, teriparatide, denosumab)
iv. Long-term oral NSAIDs: Increased risk of side effects (e.g. peptic ulcer disease, bleeding, worsening heart failure) when
taken regularly for ≥ 2 months.
v. Long-term oral corticosteroids: Increased risk of major side effects (e.g. fragility fractures, proximal myopathy, peptic
ulcer disease) when taken regularly for ≥ 2 months. Consider careful dose reduction and discontinuation.
Section H: i. Drugs for benign prostatic hyperplasia (5-alpha reductase inhibitors and alpha-blockers) in catheterized male
Urogenital patients: No benefit with longtermbladder catheterisation.
System ii. Drugs for overactive bladder (muscarinic antagonists and mirabegron): No benefit in patients with persistent, irreversible
urinary incontinenceunless clear history ofpainful detrusor hyperactivity.
Section I: i. Anti-diabetic drugs: De-intensify therapy. Avoid HbA1c targets (HbA1C <7.5% [58 mmol/mol] associated with net harm in
Endocrine this population). Goal of care is to minimize symptoms related to hyperglycaemia(e.g. excessive thirst, polyuria).
System
Section J: i. Multi-vitamin combination supplements: Discontinue when prescribed for prophylaxis rather than treatmentof hypovitaminosis.
Miscellaneous ii. Folic acid: Discontinue when treatment course completed. Usual treatment duration 1-4 months unless malabsorption,
malnutrition or concomitant methotrexate use.
iii. Nutritional supplements:Discontinue when prescribed for prophylaxis rather than treatment of malnutrition.
required, the user is not provided with resources or decision aids to estimate treat-
ment benefit–harm trade-offs in individual patients. The use of implicit medication
assessment tools is, in general, time-consuming and is likely to result in variations
in practice between physicians, and for these reasons, integration into routine clini-
cal practice has been limited [22]. Serial considerations for deprescribing according
to the CEASE algorithm of Scott et al. [21] comes in the form of four key questions
relating to any medication that the patient is taking. If the answer to these four ques-
tions in series is ‘No’, the drug may be continued. If there is a ‘Yes’ answer to any
of the four questions, deprescribing should be considered. The four questions are as
follows:
10.5 Conclusion
Larger, multi-centre, randomized trials with extended follow-up times are required
to provide further clarification on the impact of deprescribing interventions on
patient-related outcomes such as hospital admissions, quality of life and mortality.
Expectation that deprescribing interventions will improve these outcomes may be
overreaching. Deprescribing involves the withdrawal of a medical intervention,
and, therefore, demonstrating that patient’s symptoms do not subsequently deterio-
rate and that quality of life is maintained may be enough to justify the process.
The goal of deprescribing is to minimize drug-related morbidity and medication
burden for older people approaching end-of-life. Central to this goal is the recogni-
tion that clinicians have prescribing responsibilities beyond the restoration and
maintenance of health. Increasing dependency and, ultimately, death do not neces-
sarily represent a failure of medical care but are natural processes for which science
has no ultimate remedy. In this context, clinicians need to support their older frailer
10 Deprescribing: Evidence Base and Implementation 127
patients approaching end of life with personalized care that prioritizes symptom
control rather than survival extension.
Finally, deprescribing is generally acceptable to older patients and their carers
[33]. Recent evidence also indicates that deprescribing in older people is cost-
effective [34].
References
1. Woodward M. Deprescribing: achieving better health outcomes for older people through
reducing medications. J Pharm Pract Res. 2003;33:323–8.
2. Scott I, Jayathissa S. Quality of drug prescribing in older patients: is there a problem and can
we improve it? Intern Med J. 2010;40:7–18.
3. Hardy JE, Hilmer SN. Deprescribing in the last year of life. J Pharm Pract Res. 2011;41:146–51.
4. Thompson W, Farrell B. Deprescribing: what is it and what does the evidence tell us? Can J
Hosp Pharm. 2013;66:201.
5. Reeve E, Gnjidic D, Long J, Hilmer S. A systematic review of the emerging definition of
‘deprescribing’ with network analysis: implications for future research and clinical practice.
Br J Clin Pharmacol. 2015;80(6):1254–68.
6. Thillainadesan J, Gnjidic D, Green S, Hilmer SN. Impact of deprescribing interventions in
older hospitalised patients on prescribing and clinical outcomes: a systematic review of ran-
domised trials. Drugs Aging. 2018;35(4):303–19.
7. Dills H, Shah K, Messinger-Rapport B, Bradford K, Syed Q. Deprescribing medications for
chronic diseases management in primary care settings: a systematic review of randomized
controlled trials. J Am Med Dir Assoc. 2018;19(11):923–935.e2.
8. Shrestha S, Poudel A, Steadman K, Nissen L. Outcomes of deprescribing interventions in
older patients with life-limiting illness and limited life expectancy: a systematic review. Br J
Clin Pharmacol. 2019;86(10):1931–45. https://doi.org/10.1111/bcp.14113.
9. Parkes CM. Accuracy of predictions of survival in later stages of cancer. BMJ. 1972;2:29–31.
10. Christakis N, Lamont E. Extent and determinants of error in doctors’ prognoses.
BMJ. 2000;320:469–73.
11. Vigano A, Dorgan M, Buckingham J, Bruera E, Suarez-Alzamor ME. Survival predic-
tion in terminal cancer patients: a systematic review of the medical literature. Palliat Med.
2000;14:363–74.
12. Yourman LC, Lee SJ, Schonberg MA, Widera EW, Smith AK. Prognostic indices for older
adults: a systematic review. JAMA. 2012;307(2):182–92.
13. Curtin D, Dahly D, Van Smeden M, et al. Predicting 1-year mortality in older hospitalized
patients: external validation of the HOMR model. J Am Geriatr Soc. 2019;67(7):1478–83.
https://doi.org/10.1111/jgs.15958.
14. Lynn J. Living long in fragile health: the new demographics shape end of life care. Hast Cent
Rep. 2005;35(7):s14–8.
15. Downar J, Goldman R, Pinto R, Englesakis M, Adhikari NK. The “surprise question”
for predicting death in seriously ill patients: a systematic review and meta-analysis.
CMAJ. 2017;189(13):E484–93.
16. White N, Kupeli N, Vickerstaff V, Stone P. How accurate is the ‘surprise question’ at identify-
ing patients at the end of life? A systematic review and meta-analysis. BMC Med. 2017;15:139.
17. Clark D, Armstrong M, Allen A, et al. Imminence of death among hospital inpatients: preva-
lent cohort study. Palliat Med. 2014;28(6):274–9.
18. Holmes HM, Min LC, Yee M, et al. Rationalizing prescribing for older patients with multimor-
bidity: considering time to benefit. Drugs Aging. 2013;30(9):655–66.
128 D. Curtin and D. O’Mahony
19. Fortin M, Dionne J, Pinho G, Gignac J, Almirall J, Lapointe L. Randomized controlled tri-
als: do they have external validity for patients with multiple co-morbidities? Ann Fam Med.
2006;4:104–8.
20. Thompson W, Lundby C, Graabaek T, et al. Tools for deprescribing in frail older persons and
those with limited life expectancy: a systematic review. J Am Geriatr Soc. 2019;67(1):172–80.
https://doi.org/10.1111/jgs.15616.
21. Scott IA, Hilmer SN, Reeve E, et al. Reducing inappropriate polypharmacy: the process of
deprescribing. JAMA Intern Med. 2015;175(5):827–34.
22. Spinewine A, Schmader KE, Barber N, et al. Appropriate prescribing in elderly people: how
well can it be measured and optimised? Lancet. 2007;370(9582):173–84.
23. Lavan AH, Gallagher P, Parsons C, O'Mahony D. STOPPFrail (screening tool of older persons
prescriptions in frail adults with limited life expectancy): consensus validation. Age Ageing.
2017;46(4):600–7.
24. Curtin D, Gallagher P, O'Mahony D. Deprescribing in older people approaching end-of-life:
development and validation of STOPPFrail version 2. Age Ageing. 2021;50(2):465–71.
25. Lavan AH, Gallagher P, O’Mahony D. Inter-rater reliability of STOPPFrail [screening tool
of older persons prescriptions in frail adults with limited life expectancy] criteria amongst 12
physicians. Eur J Clin Pharmacol. 2018;74(3):331–8.
26. Curtin D, Dukelow T, James K, O'Donnell D, O'Mahony D, Gallagher P. Deprescribing in
multi-morbid older people with polypharmacy: agreement between STOPPFrail explicit
criteria and gold standard deprescribing using 100 standardized clinical cases. Eur J Clin
Pharmacol. 2019;75(3):427–32.
27. Potter K, Flicker L, Page A, Etherton-Beer CD. Deprescribing in frail older people: a ran-
domised controlled trial. PLoS One. 2016;11:e0149984.
28. Curtin D, Jennings E, Daunt R, et al. Deprescribing in older people approaching end of life: a
randomized controlled trial using STOPPFrail criteria. J Am Geriatr Soc. 2020;68(4):762–9.
29. Charles C, Gafni A, Whelan T. Shared decision-making in the medical encounter: what does it
mean? (Or it takes at least two to tango). Soc Sci Med. 1997;44(5):681–92.
30. Schuling J, Gebben H, Veehof LJ, Haaijer-Ruskamp FM. Deprescribing medication in very
elderly patients with multimorbidity: the view of Dutch GPs. A qualitative study. BMC Fam
Pract. 2012;13:56.
31. Anthierens S, Tansens A, Petrovic M, Christiaens T. Qualitative insights into general practitio-
ners views on polypharmacy. BMC Fam Pract. 2010;11:65.
32. Reeve E, Wolff JL, Skehan M, Bayliss EA, Hilmer SN, Boyd CM. Assessment of attitudes
toward deprescribing in older medicare beneficiaries in the United States. JAMA Intern Med.
2018;178(12):1673–80.
33. Seewoodharry M, Khunti K, Davies MJ, Gillies C, Seidu S. Attitudes of older adults and their
carers towards de-prescribing: a systematic review. Diabet Med. 2022;39(7):e14801.
34. Romano S, Figueira D, Teixeira I, Perelman J. Deprescribing interventions among community-
dwelling older adults: a systematic review of economic evaluations. PharmacoEconomics.
2022;40(3):269–95.
Part II
Geriatric Syndromes and Common Chronic
Conditions
General Principles of Management
of Patients with Multimorbidity 11
and Frailty
The progressive ageing of the population has led to a new health scenario, where
frailty and multimorbidity are extremely common. Although both are strongly asso-
ciated with poor quality of life, increased risk of disability, hospitalization, institu-
tionalization and higher mortality, multimorbidity and frailty should be considered
as different entities.
11.1.1 Multimorbidity
C. Cocchi (*)
Università Cattolica del Sacro Cuore, Rome, Italy
Fondazione Universitaria Policlinico Gemelli, IRCCS, Rome, Italy
e-mail: [email protected]
G. Onder · M. B. Zazzara
Fondazione Universitaria Policlinico Gemelli, IRCCS, Rome, Italy
of life, frequency of healthcare use, level of required assistance and survival, with a
consequent challenging medical and care management.
Multimorbidity should not be confused with comorbidity, which refers to the
presence of one or more additional conditions co-occurring with a primary condi-
tion. The difference between the two is important, because it implies a different
clinical approach. In fact, whereas multimorbidity suggests a holistic assessment
that takes into consideration the individual’s clinical complexity, equally weighting
all diseases, comorbidity implies the presence of a prevailing clinical condition
requiring attention. To put it simple, multimorbidity refers to a patient-centred
vision, while comorbidity is more disease-centred.
Even though multimorbidity as a concept is now well known and established, it
is not free from limitations. In particular, multimorbidity suggests that diseases are
equally weighted, whereas the impact on the individual’s health is greater than the
mere sum of the singular clinical effects of every disease [1, 2].
11.1.2 Polypharmacy
11.1.3 Frailty
chronic diseases has a central role in initiating or worsening frailty. In addition, the
frailty index proposed by Rockwood et al., which is based on a combination of dif-
ferent deficits, primarily includes chronic diseases as a criterion [4, 5]. In a recent
meta-analysis, the prevalence of multimorbidity in frail individuals was 72%, and
the prevalence of frailty among multimorbid individuals was 16%. This means that
most frail individuals are also multimorbid, whereas very few people with multi-
morbidity are also frail. In the general adult population, only 6% are both frail and
multimorbid [1].
As chronic diseases and multimorbidity have such a strong role in determining
frailty, it could be hypothesized that treating these conditions may counteract the
development of frailty, eventually reducing any associated negative consequences.
However, the hypothesis that intensive treatment of chronic diseases might reduce
the progression of frailty is poorly supported by existing studies. In contrast, some
evidence suggests that intensive treatment of chronic diseases may increase nega-
tive health outcomes in frail older adults. For example, polypharmacy, anticholiner-
gic and psychotropic medications have been associated with incident frailty and
pre-frailty over 2 to 5-year follow-up periods [6].
The clinical picture of patients with chronic diseases is strongly influenced by
the presence of frailty features (see Table 11.1): presence of chronic disease might
represent a limited part of the complexity of frail patients that usually present with
multiple impairments in systems and organs, and this complexity might influence
the approach to pharmacological treatment.
Table 11.1 Clinical picture of frail and non-frail patients with chronic conditionsa
Characteristic/
Domain Non-frail Frail
Chronic Diabetes, hypertension Diabetes, hypertension
diseases
Symptoms None Present (i.e. pain, dyspnoea, dizziness)
Functional Physically active Slow walking speed
status Independent in instrumental activities Need help in instrumental activities of
of daily, including managing finances daily living, including managing
and/or medications, use of finances and/or medications, use of
transportation, housework, shopping, transportation, housework, shopping,
walking outside alone, preparing walking outside alone, preparing
meals meals
Cognition Not impaired Mild to moderate impairment
Social function Availability social support (i.e. lives Poor social support (i.e. lives alone,
with spouse or relatives) limited support available from
relatives or friends)
Continence Continent Urinary incontinence
Nutrition Normal nutritional status Obesity or malnutrition, recent weight
loss
Mood Normal Depressive symptoms, sad or
depressed
a
Tables 1 and 2 inspired by Onder G. (coauthor of this work), Vetrano DL et al. “Accounting for
frailty when treating chronic diseases”. Consent obtained by other coauthors via email
134 C. Cocchi et al.
With increasing multimorbidity and frailty, the world of healthcare must reinvent
itself to find effective ways of dealing with complexity. As of today, medicine is still
single-disease oriented, an approach that appears obsolete when confronted with
today’s reality.
One of the main problems for multimorbid patients is represented by the frag-
mentation of care. Patients are often subjected to a large number of specialist visits
and treated with a large number of drugs. The models of care must be modified
to cope with this new type of patient. A new clinical approach is needed, and it
must be multidisciplinary-centred and focused on promoting individual’s
independence.
The complexity and heterogeneity of patients with multimorbidity and poly-
pharmacy make traditional guidelines often inadequate and not supportive in
decision-making. An increasing number of studies have pointed out that disease-
specific guidelines are difficult to apply in multimorbid patients. This difficulty
arises primarily from how these guidelines are developed. Indeed, disease-specific
guidelines are developed on data collected with randomized clinical trials that
exclude fragile individuals a priori, to avoid confounding factors. Second, these
guidelines often do not account for other coexisting chronic diseases: drug–drug or
drug–disease interactions may easily occur in persons suffering from multiple
chronic conditions.
New guidelines have recently been developed (i.e. Ariadne principles, NICE,
JA-CHRODIS) for the management of the multimorbid patient. The first purpose
of these guidelines is to maximize the quality of life, focusing not on the symp-
tom nor the disease but on the individual as a whole. Those guidelines present
several commonalities. First, they try to identify a target population that can
benefit from the application of these principles (e.g. fragile individuals). In addi-
tion, they support the flexible use of existing guidelines on individual diseases,
focusing on treatment burden. Moreover, they theorize the importance of coordi-
nating all the different specialists involved in patient care, often via the special-
ized figure of the case manager, who acts as facilitator. Those guidelines also
focus on a shared decision-making with the patient, in order to agree on an indi-
vidualized care plan. Lastly, they emphasize on patient and caregivers’ education
to improve self-management and self-efficacy. Below, we will analyze in more
detail the main points of the recent guidelines on the management of the multi-
morbid patient [6, 9].
11 General Principles of Management of Patients with Multimorbidity and Frailty 135
Patients with multimorbidity are heterogeneous, and their global health status and
risk of negative outcomes may vary largely. Furthermore, the consequences of mul-
timorbidity cannot be linearly estimated on the basis of the number of clinical con-
ditions. In order to develop the best care plan for each individual patient, it is
important to be able to identify different care needs. Indeed, there are older adults
who, despite the presence of multiple coexisting chronic conditions, are in good
health and have a good life expectancy, so they must receive the standard level of
assistance, including preventive programs and appropriate diagnostic-therapeutic
interventions. On the other hand, there are patients who are more at risk of develop-
ing negative events and who, therefore, need specific treatment plans.
In the population with multimorbidity, stratification strategies carried out on the
basis of patterns of different diseases, presence of symptoms, and physical, cogni-
tive, and socio-economic status help to identify the most demanding and complex-
to-treat groups—those that might benefit most from individualized and integrated
healthcare plans. In this context, the assessment of frailty with appropriate instru-
ments (i.e. Frailty index, Clinical Frailty Scale, physical performance measures) can
be used to identify the most care-demanding groups that might benefit most from
individualized and integrated approaches. Both guidelines from the National
Institute for Health and Care Excellence on multimorbidity and Italian Guidelines
on Multimorbidity and Polypharmacy suggest to assess frailty status to identify
people with multimorbidity who are susceptible and at risk of negative health out-
comes, thus more likely to benefit from a patient-centred approach [3, 10, 11].
–– Diseases and health problems, including clinical and functional status, cognition,
geriatric syndromes, life expectancy, presence of any symptoms, their severity
and their impact on quality of life
136 C. Cocchi et al.
–– Psychosocial context
–– Treatment burden (pharmacological and nonpharmacological): evaluating poten-
tial interactions or adverse effect, adherence or underuse and treatment burden
All of these elements should be reviewed regularly, with special attention for
signs and symptoms of cognitive impairment and psychological or social problems,
since they could modify our intervention [3, 6, 10–13].
Since the management of the multimorbid patient is complex and challenging, the
identification of health priorities, shared between patients and care provider, repre-
sents a relevant step to provide individualized, patient-centred care. Decisions about
the patient’s health must be made in full agreement with patient and caregivers. It is
necessary to explain the purpose of the therapeutic approach and the realistically
achievable goals so that the patient can express his/her preferences based on per-
sonal values and priorities. When suggesting a treatment, the person’s prognosis
should always be taken into consideration, as well as the patient’s attitude towards
the treatment, his expectations and objectives. Monitoring and re-discussing priori-
ties is fundamental, as they can change overtime [3, 6, 10, 11].
11.2.5 Self-management
Multimorbid patients are the major users of healthcare resources, and their involve-
ment in disease management is a goal to be pursued. Self-management of chronic
diseases is a treatment process characterized by the responsible and proactive par-
ticipation of the patient himself, allowing the patient to be aware of his own health
condition and empowering his/her involvement in the therapeutic decision-making
process. Self-management represents a fundamental lever for the effectiveness and
efficiency of the health system. On the assumption that knowledgeable and expert
patients are able to manage their quality of life to the fullest of their potential, edu-
cational programs are useful to enable patients to acquire a central role in the man-
agement of his own health. Many patients who have lived with chronic diseases for
years can be trained to manage the disease and take active responsibility of both non
pharmaceutical (i.e. diet, physical exercises) and pharmaceutical (i.e. use of medi-
cations) management [3, 6, 10, 11].
Patients with multimorbidity often receive disjointed care, which leads to ineffi-
cient, ineffective and possibly harmful clinical interventions. Ideally, care for
patients with multimorbidity should involve numerous healthcare professionals,
include various dimensions (clinical, functional and social), and be based on solid
evidence. A careful coordination by a case manager of the various healthcare pro-
fessionals is therefore essential.
Care plans for multimorbid patients are not static, but they can often change due
to a change in the patient’s priorities, to the onset of new problems or as a result of
co-occurring events. This is why it is important to re-evaluate the patient, especially
after hospitalizations or the prescription of new drugs [3, 6, 10, 11].
As mentioned earlier, the identification of frailty is a key step to stratify the popula-
tion with multimorbidity and to identify patients that need an individualized care
approach. In particular, the use of symptomatic and preventive drugs in persons with
frailty should be evaluated carefully based on several considerations.
Symptoms related to chronic diseases might have a relevant role in the onset or
worsening of frailty status, and the treatment of these symptoms might potentially
reverse frailty. For example, pain is a strong determinant of frailty in patients with
138 C. Cocchi et al.
Treatment of chronic diseases might have an impact on the onset of frailty by pre-
venting the occurrence of negative health events. For example, it would be reason-
able to hypothesize that pharmacological treatment of frail persons who experienced
a bone fracture may prevent the onset of new fractures and reduce the risk of frailty
and disability. However, evidence has suggested lack of benefit from treatment with
bisphosphonates on fractures and mortality in frail women in nursing home.
Similarly, possible benefits related to preventive pharmacological treatment of
hypertension on frailty onset are still being discussed. In observational studies,
hypertension does not seem to have an impact on frailty, and the negative effects of
antihypertensive treatment seem to be more pronounced in patients with frailty. In
addition, the negative effects of hypertension on health outcomes might be reduced
in the presence of frailty, suggesting a reverse epidemiology phenomenon in patients
with frailty.
The example of hypertension could also apply to other chronic diseases, where
the pros and the cons of intensive pharmacological treatment in frailty are still
debated, and any possible positive effects might be counterbalanced by an increased
risk of negative effects. Indeed, treatment of chronic diseases in frail individuals
may raise several concerns that might alter the risk/benefit ratio of a given treatment
(Table 11.2).
11 General Principles of Management of Patients with Multimorbidity and Frailty 139
Table 11.2 Factors challenging pharmacological treatment of chronic diseases in the presence of
frailtya
Problem Consequences Potential solutions
Exclusion of frail Uncertainties about the Inclusion of frail patients in RCTs;
persons from studies efficacy and safety of Evidence from observational studies
treatment in frail persons
Reduced life expectancy Reduced/absent Assess life expectancy;
in frail persons beneficial effects Evaluate risk-benefit ratio of treatments
Increased susceptibility Higher rate of adverse Periodic therapeutic review and
to iatrogenic events due drug events reconciliation;
to intensive treatment Evaluate risk-benefit ratio of treatments;
Focus treatment on specific goals
Functional deficits Poor medication Assess the capacity of patients to
associated with frailty adherence; self-manage their medication regimen;
Medication errors Focus treatment on health priorities;
Adjust communication strategies;
Use of tools to improve adherence;
Reduce number medications and
medication regimen complexity
(frequency of medication
administrations, use of appropriate dose
forms)
a
Tables 1 and 2 inspired by Onder G. (coauthor of this work), Vetrano DL et al. “Accounting for
frailty when treating chronic diseases”. Consent obtained by other coauthors via email
Exclusion from studies. Frail older people are often excluded from randomized
clinical trials assessing the effects of treatments for chronic diseases. Practical limi-
tation—difficulties in reaching the research centre—or and higher mortality rates
that prevent having appropriate follow-up times may in part justify this exclusion.
However, this limits the generalizability of clinical trial findings and estimation of
the efficacy and safety of treatments for chronic diseases in persons with frailty.
Limited life expectancy. Frailty is associated with limited life expectancy; esti-
mates from the SHARE study suggests that life expectancy for frail individuals at
age of 70 years ranges between 0.1 and 1.8 years in men and between 0.4 and
5.5 years in women. This suggests that several preventive treatments for chronic
disease might have limited benefits in persons with frailty, given that the time-until-
benefit might exceed the actual life expectancy in frail individuals. In frail popula-
tions, it is essential to assess life expectancy, evaluate the risk/benefit ratio of
treatments and consider discontinuing medications aimed at prolonging life or pre-
venting adverse clinical events if the time needed to obtain the expected benefits is
longer than the estimated life expectancy and when they do not offer symptomatic
benefits.
Increased risk of negative outcomes from intensive treatment. Frailty is associ-
ated with an increased risk of negative events associated with pharmacological
treatments. Some of the current guidelines for the treatment of chronic diseases
140 C. Cocchi et al.
acknowledge this increased risk of negative events. For example, guidelines for
treatment of type II diabetes supports the adoption of less strict blood glucose tar-
gets in frail patients, given the increased risk of hypoglycaemia without accruing
meaningful benefit for frail older adults.
Ability to adhere. Functional deficits related to frailty might be associated with
unintentional non-adherence through reduced ability to manage pill containers,
open child-resistant containers and/or split tablets. Functional impairment may
reduce the ability to adhere to a complex medication regimen and in case of poly-
pharmacy, increase the risk of medication errors and limit the capacity to self-report
adverse drug events. Treatment of chronic diseases in frailty should be simplified
and focused on clinical priorities, established by the patient and his/her physician.
Communication strategies should be implemented to empower and educate frail
patients and their caregivers on how to properly take a medication, discussing with
patients and caregivers about the importance of medication adherence, providing
information about health and associated treatments and suggesting the use of aids
(i.e. pill boxes) to improve adherence. Medication regimen complexity can often be
reduced through consolidating the number of dose times and using long-acting for-
mulations where appropriate [14].
environment, education and social capital, lifestyle) represent key elements of care
in the approach to multimorbidity and can influence treatment adherence and suc-
cess. This approach focuses on identifying multiple targetable risk factors of multi-
morbidity and individual pathways that could drive effective interventions and
tailored management of the persons with multimorbidity, in line with the principles
of precision medicine.
At the same time, however, numerous different individual pathways may lead to
the development of different multimorbidity phenotypes, characterized by several
combinations of individual factors. A clear understanding interplay and interaction
between these factors might be extremely complex, making the definition of indi-
vidual and personalized interventions challenging. Given multimorbidity heteroge-
neity, a possible solution to simplify the application of precision medicine to this
condition is to reduce the dimensionality of its complexity and to find homogeneous
patterns that may work as effective targets of preventive and therapeutic strategies.
Focusing on clusters of chronic diseases could represent a useful strategy aiming to
identify diseases that co-occur in the same person beyond chance. Diseases tend to
cluster together because they share common risk factors and pathophysiological
mechanisms or because one disease is the direct cause of another. Knowing how and
why diseases cluster together can lead to the development of preventive and thera-
peutic strategies targeting specific but common pathways. At the time of writing,
applying precision medicine to specific clusters of diseases remains a work in prog-
ress that requires further research including the analyses of large epidemiological
and biological datasets investigating the interplay between genetic disease predis-
position and personal and environmental determinants of multimorbidity such as
gender, social and living factors, performance status, education and lifestyles. More
information is needed to identify specific factor or combination of factors that need
to be systematically assessed in individuals with specific clusters of diseases and
understand how these factors also interact at an individual level. This approach
could help define interventions that might comprehensively target multimorbid-
ity [15].
References
1. Vetrano DL, Palmer K, Marengoni A, Marzetti E, Lattanzio F, Roller-Wirnsberger R, et al.
Frailty and multimorbidity: a systematic review and meta-analysis. J Gerontol A Biol Sci
Med Sci [Internet]. 2019 [cited 2022 Mar 15];74(5):659–666. https://pubmed.ncbi.nlm.nih.
gov/29726918/.
2. Cesari M, Pérez-Zepeda MU, Marzetti E. Frailty and multimorbidity: different ways of think-
ing about geriatrics. J Am Med Dir Assoc [Internet]. 2017 [cited 2022 Mar 16];18(4):361–4.
http://www.jamda.com/article/S1525861017300348/fulltext.
3. Onder G, Vetrano DL, Palmer K, Trevisan C, Amato L, Berti F, Campomori A, Catalano L,
Corsonello A, Kruger P, Medea G, Nobili A, Trifirò G, Vecchi S, Veronese N, Marengoni
A. Italian guidelines on management of persons with multimorbidity and polypharmacy.
[Internet]. Aging Clin Exp Res. 2022. [cited 2022 Mar 15]. https://www.sigg.it/news/
linea-guida-i nter-s ocietaria-p er-l a-g estione-d ella-m ultimorbilita-e -p olifarmacoterapia-
2021/.
142 C. Cocchi et al.
4. Buta BJ, Walston JD, Godino JG, Park M, Kalyani RR, Xue QL, et al. Frailty assessment
instruments: systematic characterization of the uses and contexts of highly-cited instruments.
Ageing Res Rev [Internet]. 2016 [cited 2022 Mar 16];26:53. /pmc/articles/PMC4806795/.
5. Clegg A, Young J, Iliffe S, Rikkert MO, Rockwood K. Frailty in elderly people. Lancet
[Internet]. 2013 [cited 2022 Mar 16];381(9868):752–62. http://www.thelancet.com/article/
S0140673612621679/fulltext.
6. Muth C, Blom JW, Smith SM, Johnell K, Gonzalez-Gonzalez AI, Nguyen TS, et al. Evidence
supporting the best clinical management of patients with multimorbidity and polypharmacy:
a systematic guideline review and expert consensus. J Intern Med [Internet]. 2019 [cited 2022
Mar 15];285(3):272–88. https://onlinelibrary.wiley.com/doi/full/10.1111/joim.12842.
7. Villacampa-Fernández P, Navarro-Pardo E, Tarín JJ, Cano A. Frailty and multimorbidity: two
related yet different concepts. Maturitas [Internet]. 2017 [cited 2022 Mar 15];95:31–5. https://
pubmed.ncbi.nlm.nih.gov/27889050/.
8. Zucchelli A, Vetrano DL, Marengoni A, Grande G, Romanelli G, Calderón-Larrañaga A,
et al. Frailty predicts short-term survival even in older adults without multimorbidity. Eur
J Intern Med [Internet]. 2018 [cited 2022 Mar 15];56:53–6. http://www.ejinme.com/article/
S0953620518302620/fulltext.
9. Yarnall AJ, Sayer AA, Clegg A, Rockwood K, Parker S, Hindle JV. New horizons in multimor-
bidity in older adults. Age Ageing [Internet]. 2017 [cited 2022 Mar 15];46(6):882–8. https://
academic.oup.com/ageing/article/46/6/882/4103436.
10. Overview | Multimorbidity: clinical assessment and management | Guidance | NICE [Internet].
[cited 2022 Mar 19]. https://www.nice.org.uk/guidance/ng56.
11. Muth C, van den Akker M, Blom JW, Mallen CD, Rochon J, Schellevis FG, et al. The
Ariadne principles: how to handle multimorbidity in primary care consultations. BMC Med
[Internet]. 2014 [cited 2022 Mar 19];12(1):1–11. https://bmcmedicine.biomedcentral.com/
articles/10.1186/s12916-014-0223-1
12. Boyd C, Smith CD, Masoudi FA, Blaum CS, Dodson JA, Green AR, et al. Decision mak-
ing for older adults with multiple chronic conditions: executive summary for the American
Geriatrics Society guiding principles on the care of older adults with multimorbidity. J Am
Geriatr Soc [Internet]. 2019 [cited 2022 Mar 19];67(4):665–73. https://onlinelibrary.wiley.
com/doi/full/10.1111/jgs.15809.
13. Mazya AL, Garvin P, Ekdahl AW. Outpatient comprehensive geriatric assessment: effects on
frailty and mortality in old people with multimorbidity and high health care utilization. Aging
Clin Exp Res [Internet]. 2019 [cited 2022 Mar 15];31(4):519–25. https://pubmed.ncbi.nlm.
nih.gov/30039453/.
14. Onder G, Vetrano DL, Marengoni A, Bell JS, Johnell K, Palmer K. Accounting for frailty when
treating chronic diseases. Eur J Intern Med [Internet]. 2018 [cited 2022 Mar 15];56:49–52.
http://www.ejinme.com/article/S0953620518300815/fulltext.
15. Onder G, Bernabei R, Vetrano DL, Palmer K, Marengoni A. Facing multimorbidity in the
precision medicine era. Mech Ageing Dev. 2020;190:111287.
Frailty and Drug Therapy
12
Annette Eidam, Matteo Cesari, and Jürgen M. Bauer
–– The physical phenotype model. Proposed in 2001 by Fried and colleagues [4],
this model is based on the evaluation of five signs/symptoms (i.e., exhaustion,
slowness, muscle weakness, involuntary weight loss, sedentary behavior) pheno-
typically describing the frail individual. Older adults who are negative for all
these criteria are considered robust, those who are positive for one or two are
considered prefrail, and those who are positive for three or more are consid-
ered frail.
–– The accumulation of health deficit model. This model was proposed by Mitnitski
and colleagues in 2001 [5]. It stems from the observation that the aging process
However, there is still an ongoing discussion on the best method for diagnosing
frailty, in clinical practice and in research as well [6]. Besides the physical pheno-
type and the Frailty Index, a large number of other instruments to assess frailty have
been developed and are used, with the exception of the Clinical Frailty Scale mostly
in research settings. Examples include the Edmonton Frail Scale and the Groningen
Frailty Indicator [7].
Independently of the preferred model for its operationalization, frailty has been
acknowledged by the scientific community not only as a clinically relevant condi-
tion associated with major adverse health outcomes (e.g., hospitalization, institu-
tionalization, disability, falls, death [8]) but also as a valuable approach that can
improve the care of older persons [9]. In fact, it has become evident that chronologi-
cal age does not adequately capture the heterogeneity of the biological, clinical, and
social background of older persons [10]. Furthermore, a preference of chronological
age over biological age in the clinical decisional process may increase ageism in
medicine by excluding older persons from preventive and therapeutic options. In
other words, frailty may represent an alternative approach for measuring the indi-
vidual’s biological age.
The detection of frailty and the exploration of its underlying causes are critical
for the proper management of the older person. It means understanding whether the
individual, regardless of his/her age, is biologically fit to sustain and benefit from
standard interventions or whether an adapted approach with potentially different
treatment strategies and objectives should be put in place. The adapted approach
may include the design of a personalized plan aimed at solving potentially revers-
ible barriers to restore (at least, partially) robustness to allocate the highest stan-
dards of care. Such an adapted approach should also acknowledge the particular
vulnerabilities of the frail older patient.
In this context, the evaluation and management of frailty play an important
role in the drugs prescription. The holistic approach needed to identify the under-
lying causes of frailty (i.e., the comprehensive geriatric assessment) allows to
measure the priorities, needs, and resources of the individual [8]. This approach
supports the clinician in the therapeutical choice by allowing a more informed
evaluation of the risk–benefit profile of their frail patient. However, it cannot be
overlooked that frail older persons are often excluded from randomized con-
trolled trials given their complexity [11]. For this reason, the conduction of evi-
dence-based medicine and prescribing in this population are frequently
challenging. Frailty may, thus, become a condition to weigh the endogenous and
exogenous factors potentially interfering with the “one-fits-all” approach typical
12 Frailty and Drug Therapy 145
in this population compared to estimations based on cystatin C [25, 26]. There has
been emerging evidence that the difference between estimated GFR based on cys-
tatin C and estimated GFR based on creatinine might suggest the presence of frailty
[27, 28]. Given the high number of renally eliminated drugs used by frail older
patients, careful dose adaptation appears to be particularly relevant in this popula-
tion [13, 25].
To date, only a limited number of pharmacokinetic studies have used validated
frailty assessments to characterize their study populations [15]. So far, Fried and
colleagues’ physical phenotype of frailty [22–24] and variants of the Edmonton
Frail Scale [18, 26, 29, 30] remain the most frequently used assessment instruments
in this context. A small number of studies have used disease-specific assessments
(International Myeloma Working Group Frailty Score [31]) or functional parame-
ters (grip strength [26]) to evaluate the presence of frailty. Applying a standardized
assessment to measure frailty would be essential for future pharmacokinetic studies,
as this approach would facilitate the comparison across studies [15]. At present,
many instruments are used to diagnose frailty, which differ significantly in their
components and consecutively in the populations that are interpreted as being frail
[7, 32]. It remains to be identified which frailty criteria would be particularly rele-
vant to pharmacokinetics in older persons and could support optimized dosing
schemes in this patient group.
Age-sensitive eligibility criteria often exclude older adults from clinical drug trials
[33]. A recent evaluation found that almost 80 % of adults with elevated blood pres-
sure aged ≥ 80 years met at least one exclusion criterion of two major clinical trials
that informed guideline recommendations for arterial hypertension [34]. Frail older
adults are particularly underrepresented in clinical trials [35]. Moreover, trials that
include a relevant number of older adults usually do not assess and report on the
participants’ frailty status [35]. As frailty is associated with both mortality [36] and
adverse drug reactions [14], the therapeutic outcome and especially the risk–benefit
ratio of drug therapy might differ significantly in frail compared to robust older
persons. Therefore, it is essential to include older adults in clinical drug trials and
characterize older study populations regarding their functionality, including frailty.
In the absence of standardized baseline frailty assessments of clinical trial popu-
lations, several research groups have attempted to identify frail study participants
retrospectively. By including a 26-item frailty index in a pooled analysis of 14 mul-
ticenter trials of primarily cardiovascular medicines, Farooqi and colleagues found
that frail participants had a higher risk of cardiovascular events, cardiovascular mor-
tality, and all-cause mortality than non-frail participants [37]. Using a 40-item
frailty index, Hanlon and colleagues reported that mild to moderate frailty was
unexpectedly common in phase 3/4 trials of drug therapies for rheumatoid arthritis,
type 2 diabetes mellitus, and chronic obstructive pulmonary disease [38]. Brefka
and colleagues proposed a categorization for the retrospective identification of older
12 Frailty and Drug Therapy 147
fibrillation and a score ≥ 3 on the Groningen Frailty Indicator [46]. Few studies
focus on frailty as an outcome parameter of drug therapy. The phase 2 trial
“Metformin for Preventing Frailty in High-risk Older Adults” (https://clinicaltrials.
gov/ct2/show/NCT02570672) investigates whether regular intake of metformin
could prevent the progression of frailty status in robust and prefrail older adults with
prediabetes [47]. In this trial, Fried and colleagues’ physical phenotype is used.
12.7.1 Deprescribing
There has been a growing interest in the concept of deprescribing and in withdraw-
ing potentially harmful and/or unnecessary medicines in older adults, particularly in
those with frailty. Current research focuses on the question, whether deprescribing
might be feasible without causing a negative impact on favorable health outcomes
and whether it might reduce adverse drug reactions that contribute to frailty.
Several instruments have been developed to address deprescribing in frail older
adults [83]. Such instruments may offer a framework for targeting deprescribing, a
deprescribing approach for the patient’s entire medication list, or guidance on
deprescribing specific medications [83]. Some deprescribing instruments have been
developed for a particular subgroup within the frail population. For example, in
2017, Lavan and colleagues published the Screening Tool of Older Persons
Prescriptions in Frail adults with limited life expectancy (STOPPFrail) [84]. They
later provided an updated version of this tool [85]. STOPPFrail has been designed
to support physicians with deprescribing medications in older patients living with
severe frailty who approach the end of their lives. This tool, therefore, only applies
to this specific group of frail individuals. It does not apply to all frail patients and
especially not to prefrail individuals. The tool was validated using the Delphi con-
sensus methodology [84, 85], and currently includes 25 deprescribing criteria [85].
The evidence from empirical studies examining the practicality and the benefits
and harms of deprescribing in older patients with frailty is still limited, and com-
parison of deprescribing studies in frail older individuals is complicated due to het-
erogeneity in interventions, outcome measures, and frailty criteria [86]. Nevertheless,
though still incomplete, existing research indicates that deprescribing in this patient
group might be both feasible and safe as well as beneficial [86].
Frailty is not only associated with polypharmacy and the prescription of potentially
harmful medicines but also with potential prescribing omissions and underprescrip-
tion of potentially beneficial medicines. The individual frailty status may affect the
decision whether to start or to withhold anticoagulation therapy in older adults with
atrial fibrillation [87, 88]. A retrospective cohort study found that between 2009 and
2018, the prescription of anticoagulants had increased in patients with nonvalvular
atrial fibrillation who met guideline indications for anticoagulation [89]. However,
this increase was less pronounced in the frail population [89]. Initiation of statin
treatment after acute coronary syndrome was inversely correlated to the degree of
frailty in older patients [90]. In a prospective population-based cohort study in older
adults, underuse of preventive cardiovascular medicines was significantly associ-
ated with frailty [91].
152 A. Eidam et al.
Network has named international consensus principles for research activities in this
context. The authors emphasize the need to include frail older adults in randomized
controlled trials and identify strategies that would improve recruitment and reduce
dropout rates in this population [94]. Research studies should include outcome mea-
sures that reflect the priorities of frail older patients by focusing both on physical
health and on social well-being, such as physical and cognitive function as well as
functional independence [94]. Moreover, the authors encourage research in the
nursing home setting, which has been particularly underrepresented in clinical stud-
ies despite the fact that nursing home residents carry a notably high medication
burden [94].
Frailty might be a better predictor of an individual’s response to drug therapy than
chronological age, and frailty is likely to modify the effect and risk–benefit profile of
medicines [94, 98]. However, reflecting the lack of an international consensus on the
diagnostic criteria of frailty, it is still unclear which frailty assessment could be best
suited for application in drug trials and for evaluating the effects of polypharmacy in
the older population. Although different frailty measures correlate significantly with
each other, different frail populations are identified by the various assessment tools
[99, 100]. However, the best choice might differ between study settings, as the physi-
cal, cognitive, emotional, or social domains of the frailty construct are likely to be of
varying relevance to pharmacokinetic studies, randomized controlled drug trials, or
studies examining medication management strategies. Furthermore, the choice of the
instrument for the detection of frailty should also be made considering the interven-
tion and resources that will be allocated if the screening result is positive.
Frail older adults carry an exceptionally high medication burden. Without com-
prehensive evidence on the benefits and the risks of drug therapy in this patient
group, prescribing in frail older individuals is likely to be inadequate and potentially
harmful. Academic researchers and regulatory authorities are called upon to design
feasible strategies to recruit and characterize frail older individuals in pharmaco-
logical research, and all clinicians involved in the medical care of older patients
need to acknowledge the demands of this important population during the prescrib-
ing process.
References
1. Chen X, Mao G, Leng SX. Frailty syndrome: an overview. Clin Interv Aging. 2014;9:433–41.
https://doi.org/10.2147/CIA.S45300.
2. Morley JE, Vellas B, van Kan GA, Anker SD, Bauer JM, Bernabei R, et al. Frailty con-
sensus: a call to action. J Am Med Dir Assoc. 2013;14(6):392–7. https://doi.org/10.1016/j.
jamda.2013.03.022.
3. Cesari M, Gambassi G, van Kan GA, Vellas B. The frailty phenotype and the frailty index:
different instruments for different purposes. Age Ageing. 2014;43(1):10–2. https://doi.
org/10.1093/ageing/aft160.
4. Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, et al. Frailty in older
adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci. 2001;56(3):M146–56.
https://doi.org/10.1093/gerona/56.3.m146.
154 A. Eidam et al.
24. Hubbard RE, O’Mahony MS, Calver BL, Woodhouse KW. Plasma esterases and inflamma-
tion in ageing and frailty. Eur J Clin Pharmacol. 2008;64(9):895–900. https://doi.org/10.1007/
s00228-008-0499-1.
25. Ballew SH, Chen Y, Daya NR, Godino JG, Windham BG, McAdams-DeMarco M, et al.
Frailty, kidney function, and polypharmacy: the atherosclerosis risk in communities (ARIC)
study. Am J Kidney Dis. 2017;69(2):228–36. https://doi.org/10.1053/j.ajkd.2016.08.034.
26. Tan SJ, Cockcroft M, Page-Sharp M, Arendts G, Davis TME, Moore BR, et al. Population
pharmacokinetic study of ceftriaxone in elderly patients, using cystatin C-based estimates of
renal function to account for frailty. Antimicrob Agents Chemother. 2020;64(10):e00874–20.
https://doi.org/10.1128/AAC.00874-20.
27. Potok OA, Ix JH, Shlipak MG, Katz R, Hawfield AT, Rocco MV, et al. The difference between
cystatin C- and creatinine-based estimated GFR and associations with frailty and adverse
outcomes: a cohort analysis of the systolic blood pressure intervention trial (SPRINT). Am J
Kidney Dis. 2020;76(6):765–74. https://doi.org/10.1053/j.ajkd.2020.05.017.
28. Potok OA, Katz R, Bansal N, Siscovick DS, Odden MC, Ix JH, et al. The difference between
cystatin C- and creatinine-based estimated GFR and incident frailty: an analysis of the cardio-
vascular health study (CHS). Am J Kidney Dis. 2020;76(6):896–8. https://doi.org/10.1053/j.
ajkd.2020.05.018.
29. Johnston C, Hilmer SN, McLachlan AJ, Matthews ST, Carroll PR, Kirkpatrick CM. The
impact of frailty on pharmacokinetics in older people: using gentamicin population pharma-
cokinetic modeling to investigate changes in renal drug clearance by glomerular filtration.
Eur J Clin Pharmacol. 2014;70(5):549–55. https://doi.org/10.1007/s00228-014-1652-7.
30. Hias J, Van der Linden L, Walgraeve K, Gijsen M, Mian P, Koch BCP, et al. Pharmacokinetics
of 2 oral paracetamol formulations in hospitalized octogenarians. Br J Clin Pharmacol.
2022;88:1020–30. https://doi.org/10.1111/bcp.15049.
31. Nampoothiri RV, Kasudhan KS, Patil AN, Malhotra P, Khadwal A, Prakash G, et al. Impact
of frailty, melphalan pharmacokinetics, and pharmacogenetics on outcomes post autolo-
gous hematopoietic cell transplantation for multiple myeloma. Bone Marrow Transplant.
2019;54(12):2088–95. https://doi.org/10.1038/s41409-019-0631-0.
32. Faller JW, Pereira DDN, de Souza S, Nampo FK, Orlandi FS, Matumoto S. Instruments
for the detection of frailty syndrome in older adults: a systematic review. PLoS One.
2019;14(4):e0216166. https://doi.org/10.1371/journal.pone.0216166.
33. Beers E, Moerkerken DC, Leufkens HG, Egberts TC, Jansen PA. Participation of older
people in preauthorization trials of recently approved medicines. J Am Geriatr Soc.
2014;62(10):1883–90. https://doi.org/10.1111/jgs.13067.
34. Anderson TS, Odden MC, Penko J, Kazi DS, Bellows BK, Bibbins-Domingo
K. Characteristics of populations excluded from clinical trials supporting intensive blood
pressure control guidelines. J Am Heart Assoc. 2021;10(7):e019707. https://doi.org/10.1161/
JAHA.120.019707.
35. Cerreta F, Padrao A, Skibicka-Stepien I, Strampelli A, de Orbe Izquierdo MS. Medicines
for older people: assessment and transparency at the European Medicines Agency regarding
cardiovascular and antithrombotic medicinal products. Eur Geriatr Med. 2018;9(4):415–8.
https://doi.org/10.1007/s41999-018-0071-1.
36. Ekram A, Woods RL, Britt C, Espinoza S, Ernst ME, Ryan J. The association between frailty
and all-cause mortality in community-dwelling older individuals: an umbrella review. J
Frailty Aging. 2021;10(4):320–6. https://doi.org/10.14283/jfa.2021.20.
37. Farooqi MAM, Gerstein H, Yusuf S, Leong DP. Accumulation of deficits as a key risk factor
for cardiovascular morbidity and mortality: a pooled analysis of 154 000 individuals. J Am
Heart Assoc. 2020;9(3):e014686. https://doi.org/10.1161/JAHA.119.014686.
38. Hanlon P, Butterly E, Lewsey J, Siebert S, Mair FS, McAllister DA. Identifying frailty in
trials: an analysis of individual participant data from trials of novel pharmacological interven-
tions. BMC Med. 2020;18(1):309. https://doi.org/10.1186/s12916-020-01752-1.
39. Brefka S, Dallmeier D, Muhlbauer V, von Arnim CAF, Bollig C, Onder G, et al. A pro-
posal for the retrospective identification and categorization of older people with functional
156 A. Eidam et al.
86. Ibrahim K, Cox NJ, Stevenson JM, Lim S, Fraser SDS, Roberts HC. A systematic review of
the evidence for deprescribing interventions among older people living with frailty. BMC
Geriatr. 2021;21(1):258. https://doi.org/10.1186/s12877-021-02208-8.
87. Papakonstantinou PE, Asimakopoulou NI, Papadakis JA, Leventis D, Panousieris M,
Mentzantonakis G, et al. Frailty status affects the decision for long-term anticoagulation
therapy in elderly patients with atrial fibrillation. Drugs Aging. 2018;35(10):897–905. https://
doi.org/10.1007/s40266-018-0587-6.
88. Sanghai SR, Liu W, Wang W, Rongali S, Orkaby AR, Saczynski JS, et al. Prevalence of frailty
and associations with oral anticoagulant prescribing in atrial fibrillation. J Gen Intern Med.
2022;37(4):730–6. https://doi.org/10.1007/s11606-021-06834-1.
89. Orlandi M, Dover DC, Sandhu RK, Hawkins NM, Kaul P, McAlister FA. The introduc-
tion of direct oral anticoagulants has not resolved treatment gaps for frail patients with
nonvalvular atrial fibrillation. Can J Cardiol. 2022;38(1):77–84. https://doi.org/10.1016/j.
cjca.2021.09.021.
90. Lopez D, Nedkoff L, Briffa T, Preen DB, Etherton-Beer C, Flicker L, et al. Effect of frailty
on initiation of statins following incident acute coronary syndromes in patients aged >/=75
years. Maturitas. 2021;153:13–8. https://doi.org/10.1016/j.maturitas.2021.07.006.
91. Meid AD, Quinzler R, Freigofas J, Saum KU, Schottker B, Holleczek B, et al. Medication
underuse in aging outpatients with cardiovascular disease: prevalence, determinants, and
outcomes in a prospective cohort study. PLoS One. 2015;10(8):e0136339. https://doi.
org/10.1371/journal.pone.0136339.
92. Pazan F, Petrovic M, Cherubini A, Onder G, Cruz-Jentoft AJ, Denkinger M, et al. Current
evidence on the impact of medication optimization or pharmacological interventions on
frailty or aspects of frailty: a systematic review of randomized controlled trials. Eur J Clin
Pharmacol. 2021;77(1):1–12. https://doi.org/10.1007/s00228-020-02951-8.
93. Saeed D, Carter G, Parsons C. Interventions to improve medicines optimisation in frail
older patients in secondary and acute care settings: a systematic review of randomised con-
trolled trials and non-randomised studies. Int J Clin Pharm. 2022;44(1):15–26. https://doi.
org/10.1007/s11096-021-01354-8.
94. Liau SJ, Lalic S, Sluggett JK, Cesari M, Onder G, Vetrano DL, et al. Medication management
in frail older people: consensus principles for clinical practice, research, and education. J Am
Med Dir Assoc. 2021;22(1):43–9. https://doi.org/10.1016/j.jamda.2020.05.004.
95. Chen EY, Bell JS, Ilomaki J, Keen C, Corlis M, Hogan M, et al. Medication regimen com-
plexity in 8 Australian residential aged care facilities: impact of age, length of stay, comorbid-
ity, frailty, and dependence in activities of daily living. Clin Interv Aging. 2019;14:1783–95.
https://doi.org/10.2147/CIA.S216705.
96. Tinetti ME, Naik AD, Dindo L, Costello DM, Esterson J, Geda M, et al. Association of patient
priorities-aligned decision-making with patient outcomes and ambulatory health care burden
among older adults with multiple chronic conditions: a nonrandomized clinical trial. JAMA
Intern Med. 2019;179(12):1688–97. https://doi.org/10.1001/jamainternmed.2019.4235.
97. Verdoorn S, Kwint HF, Blom JW, Gussekloo J, Bouvy ML. Effects of a clinical medica-
tion review focused on personal goals, quality of life, and health problems in older per-
sons with polypharmacy: a randomised controlled trial (DREAMeR-study). PLoS Med.
2019;16(5):e1002798. https://doi.org/10.1371/journal.pmed.1002798.
98. Tan ECK, Sluggett JK, Johnell K, Onder G, Elseviers M, Morin L, et al. Research priorities
for optimizing geriatric pharmacotherapy: an international consensus. J Am Med Dir Assoc.
2018;19(3):193–9. https://doi.org/10.1016/j.jamda.2017.12.002.
99. Rockwood K, Andrew M, Mitnitski A. A comparison of two approaches to measuring
frailty in elderly people. J Gerontol A Biol Sci Med Sci. 2007;62(7):738–43. https://doi.
org/10.1093/gerona/62.7.738.
100. Roppolo M, Mulasso A, Gobbens RJ, Mosso CO, Rabaglietti E. A comparison between uni-
and multidimensional frailty measures: prevalence, functional status, and relationships with
disability. Clin Interv Aging. 2015;10:1669–78. https://doi.org/10.2147/CIA.S92328.
Falls and Impaired Mobility
13
Lotta Seppala and Nathalie van der Velde
Falls are a significant and rapidly growing global public health problem. A third of
people aged 65 years and older fall at least once per year [1]. In long-term care, falls
are even more common. Approximately, half of the nursing home residents will
experience one or more falls yearly [2]. Furthermore, in adult inpatients, falls are
the most frequently reported safety incident [3]. For example, the rate of falls has
been estimated to range from 3.3 to 11.5 falls per 1000 patient days in US hospitals
[4]. Falls have substantial impact on both mortality and morbidity. More than
684,000 people die as the result of a fall every year globally [5]. Older people are at
highest risk of dying as a result of a fall and this risk grows with advancing age [5].
Falls can have both physical and mental consequences. Over 60% of those who
experienced a fall in the past year report being injured [6]. It has been estimated that
annually, 172 million are left with short or long-term disability globally [5]. The
main physical consequences in older persons include hip fracture, other fractures,
traumatic brain injury, damage to intrathoracic and intra-abdominal organs, spinal
and nerve injuries, joint distortion and dislocation, soft-tissue damage, and bruises
and cuts [5]. Fear of falling (FOF) is common among older fallers, and FOF is
linked to impaired mobility and decreased functional status [1]. Negative mental
health effects in older persons include social withdrawal, symptom of anxiety and
depression [1]. Also, due to all negative health-related effects, falls place a
Mostly falls result from several interacting risk factors [8], of which medication use
and mobility, both modifiable key risk factors, will be discussed in detail in this
chapter. Fall risk factors have been intensively studied during the past decades. In
general, fall risk factors can be classified into intrinsic (such as medical conditions)
and extrinsic risk factors (such as home hazards) [8]. Perhaps the most important
risk factors in terms of predictive power are older age and history of past falls [5].
Some of the other key risk factors are first of all as earlier mentioned the use of
certain medications and mobility disorders as well as poor balance, visual impair-
ment, cognitive impairment and Parkinson’s disease, arthritis, and/or depression
[5]. The risk factors for falls in long-term care or in hospital differ somewhat from
the risk factors among community-dwellers [5].
Impaired mobility is a key risk factor for falls and includes impairments in muscle
strength, balance and/or gait [5]. In a systematic review by Deandrea et al., gait
problems doubled the odds of falling [9]. Also, quantitative performance measures
of gait including gait variability and gait speed have been shown to be robust predic-
tors of fall risk [10]. Gait disorders are believed to occur among a third of older
adults aged 70 years or older [11]. The origins of gait disorders include orthopedic
problems (e.g., osteoarthritis and skeletal deformities), neurological conditions
(e.g., sensory or motor impairments), and medical conditions (e.g., respiratory
insufficiency, heart failure, peripheral arterial occlusive disease, and obesity) [12],
and also certain medication use is linked to reduced gait speed [13]. With advancing
age, gait disorders usually have multiple origins [12]. Also, balance disorders are
one of the most significant reasons resulting in falls [5]. Sensory, motor, and central
processing mediate balance activity, and impairment in any of these three systems
can lead to a deficit in balance control [14]. Sensory inputs are integrated by the
central nervous system, and the central nervous system created the motor com-
mands [14]. The impairments can be due to the progressive decline of function in
the course of normal aging or due to specific pathology including use of certain
medications [14]. The prevalence of balance impairment has been estimated to vary
between 20 and 50% [14]. Last, muscle weakness (in particular lower extremity
weakness) is a major risk factor for falls [15]. Muscle strength is an essential com-
ponent of preserved mobility. The rate of muscle loss has been estimated to vary
from 1 to 2% past the age of 50 years [16]. Aging and inactivity or disuse is associ-
ated with a decline in strength, muscle mass, and structure [16]. Use of certain
medications can negatively affect either muscle strength or mass or both.
13 Falls and Impaired Mobility 163
Table 13.1 Fall risk-increasing drugs according to Delphi consensus effort [24] and their possi-
ble mechanisms leading to falls
• Benzodiazepines and benzodiazepine-related drugs
– Muscular weakness, ataxia, sedation, extrapyramidal symptoms, imbalance and/or
dizziness, visual disorders, delirium, possibly orthostatic hypotension [25, 26]
• Antipsychotics
– Sedation, drowsiness or somnolence, dizziness or vertigo, orthostatic hypotension,
extrapyramidal side effects, cardiac effects (QTc prolongation and tachycardia),
anticholinergic effects (e.g., blurred vision), delirium, confusion, hyponatremia [27, 28]
• Antidepressants
– Sedation, impaired balance/reaction time, orthostatic hypotension, cardiac conduction
and rhythm disorders, visual impairment, hyponatremia, delirium, and drug-induced
movement disorders [29]
• Diuretics
– Orthostatic hypotension, hypotension, electrolyte disturbances [30]
• Alpha-blockers used as antihypertensives
– Orthostatic hypotension, hypotension, dizziness, asthenia, and syncope [31]
• Centrally acting antihypertensives
– Orthostatic hypotension, hypotension, weakness and sedation [32]
• Vasodilators used in cardiac diseases
– Hypotension, orthostatic hypotension, dizziness, syncope [33]
• Opioids
– Orthostatic hypotension, drowsiness, sedation, confusion, delirium, eye disorders, muscle
problems (e.g., rigidity) [34]
• Antiepileptics
– Drowsiness, fatigue, dizziness, unsteadiness, vertigo, imbalance, ataxia, diplopia,
cognitive impairment, and hyponatraemia [35]
• Anticholinergics
– Effects on central nervous system (sedation, confusion, delirium, dizziness, cognitive
impairment, impaired concentration) and blurred vision, tachycardia [36]
• Alpha-blockers used for prostate hyperplasia
– Orthostatic hypotension, hypotension, dizziness, somnolence, visual impairment, and
syncope [37, 38]
• Overactive bladder and incontinence medications
– Dizziness or vertigo, somnolence, delirium, visual impairment [38, 39]
• Antihistamines
– Central nervous system side effects (e.g., sedation, drowsiness, somnolence, fatigue,
cognitive decline), anticholinergic effects, cardiovascular toxicities (e.g., arrhythmias,
prolongation of the QT interval, and postural hypotension) [40]
Table 13.3 The differences of fall risk-increasing properties between pharmacological subclasses
according to STOPPFall
Antipsychotics: agents with strong (1) sedative, (2) anticholinergic, and (3) alpha-receptor
properties
Opioids: strong opioidsa
Antidepressants: tricyclic antidepressants (TCAs), agents with strong (1) sedative effects, (2)
propensity to cause orthostatic hypotension, and (3) anticholinergic activity
Anticholinergics: agents high anticholinergic activity
Antiepileptics: older generation antiepileptics, agents with strong sedative effects
Diuretics: loop diuretics
Alpha-blockers for benign prostatic hyperplasia: nonselective alpha-blockers
Antihistamines: first-generation antihistamines, agents with strong (1) sedative effects and (2)
anticholinergic activity
Medications for overactive bladder and urge incontinence: agents with strong
anticholinergic activity
Oral hypoglycemics: agents that can cause hypoglycemia
a
A low dose of a more potent opioid, e.g., morphine, may be better tolerated than weak opioids at
a higher dose
benzodiazepine-related falls has been found to be highest after starting the therapy
[45, 46], and the association between thiazide prescription and falls was found to be
strongest in the 3 weeks after prescription [47]. The role of drug-drug interactions in
medication-related falls is still unclear [20]. But potentially, drug-drug interactions
can cause additive pharmacological effects and lead to cytochrome P-450 interactions
possibly resulting in increased drug efficacy and toxicity [48]. Furthermore, some
patient characteristics appear to be of importance when defining the medication-
related fall risk of an individual. Tinetti et al. reported that antihypertensive use was
associated with an increased risk of serious fall injuries especially among those with
history of injurious falls, which could be a marker, for example, for multimorbidity or
frailty [49]. Ham et al. showed that individuals using benzodiazepines and having
reduced CYP2C9 enzyme activity, on the basis of their genotype, had an increased fall
risk [50]. Nevertheless in general the majority of studies have focused on general risk
related to medication use instead of personalized medication optimization.
166 L. Seppala and N. van der Velde
The new world guidelines for fall prevention and management exist to guide profes-
sionals regarding fall prevention in clinical practice [51]. According to the guide-
lines, all older adults should be advised on fall prevention and physical activity [51],
and those considered at high risk should be offered a comprehensive multifactorial
fall risk assessment with a view to codesign and implement personalized multido-
main interventions [51]. One of the standard domains to be included in the interven-
tion is medication review [51]. This is supported by among others the recent network
meta-analysis showing that basic fall risk assessment including a medication review
is one the effective components of multifactorial fall prevention intervention [52].
There is however uncertainty regarding the effectiveness of deprescribing as a
single intervention in fall prevention in the literature. The meta-analysis by Lee et al.
showed no effect of pure FRID deprescribing on any of the fall outcomes as a single
intervention [53]. In addition, Cameron et al. reported that a medication review, as a
single intervention, may make little or no difference to risk of falling or rate of falls
in long-term care facilities [54]. Nevertheless, in a meta-analysis of the FRID work-
ing group of the “World Falls Guidelines for the Prevention and Management of Falls
in Older Adults” investigating the effect of medication reviews, a trend for a lower
number of fallers in long-term care was seen [55]. This suggests that in a frailer sub-
group of older adults (e.g., in case of severe frailty, advanced dementia, long-term
care residents), deprescribing might be effective also as a single intervention. Thus,
as recommended by the World Falls Guidelines Task Force, in long-term care resi-
dents, the fall prevention strategy should always include rational deprescribing of fall
risk-increasing drugs [51]. However, given the multifactorial nature of falls, depre-
scribing interventions are recommended to be implemented as a part of multimodal
strategy rather than a stand-alone strategy [55], and multifactorial fall prevention
intervention has consistently shown to reduce falls rate [56]. Common components
of multiple interventions significantly associated with a reduction in number of fall-
ers and falls rate are exercise, assistive technology, quality improvement strategies,
environmental assessment and modifications, and basic fall risk assessment (includ-
ing a medication review) as mentioned above [52]. Nevertheless, in current clinical
practice, deprescribing FRIDs after a fall incident is not frequently conducted.
According to a recent review, FRID use was not reduced at 1 and 6 months following
the fall-related healthcare contact in observational studies [17]. In addition, when
implementing fall preventive strategies among primary care providers, majority of
patients with high fall risk received most of recommended interventions and assess-
ments, except medication modification. The reluctance can be partly explained by
the general overestimation of the benefits of the medication and underestimation of
the potential harms by both physicians and patients [20]. In additions, many physi-
cians perceive the uncertainty related to the consequences of withdrawing FRIDs as
uncomfortable and challenging [20]. Nevertheless, in general, deprescribing of
FRIDs can be conducted safely [57]. Few adverse withdrawal effects occur, and if
symptoms reoccur, they can be treated safely by reintroducing the stopped medica-
tion or if possible a safer alternative [57]. To increase the implementation uptake of
13 Falls and Impaired Mobility 167
As described above, FRIDs have mostly cardiovascular, motoric, and central nervous
system related adverse effects which can lead to falls, and most of these adverse effects
are reversible after deprescribing [43, 44]. In terms of mobility outcomes, van der Velde
et al. found that withdrawal of FRIDs among geriatric outpatients improved Timed Up
and Go Test and the walking time on the 10-m walking test in a follow-up (mean,
6.7 months) and reduced occurrence of orthostatic hypotension [43, 44]. In addition,
Tsunoda et al. found that benzodiazepine withdrawal improved the stability of the body
and a recovery of cognitive functions [60]. In a study by Nurminen et al., benzodiaze-
pine withdrawal was found to improve muscle strength and balance rapidly [61].
The first step in reducing the risk of falls caused by FRIDs is to prevent their inap-
propriate use in the older population (primary prevention) [20]. Therefore, as rec-
ommended by the World Falls Guideline Task Force, a history of falls and risk of
falling should be checked before prescribing FRIDs, and the risks and benefits
should be weighted for older persons [51]. In addition, possible safer alternatives
should be considered. Furthermore, medication review (including FRIDs review)
should be performed regularly (at least annually in older persons). In frail older
adults, a medication review should be done minimally every 6 months [62–64]. In a
medication review of risks for developing adverse drug events (ADEs), the safety
and effectiveness of each drug and compliance are assessed, and views of patients
and carers are being considered [65]. FRID review is an essential part of the regular
medication review as falls are important ADEs. The regularity of these reviews will
help to keep the exposure to FRIDs as short as possible and potentially reduce asso-
ciated fall risk. It is recommended to use a validated, structured screening and
assessment tool (such as STOPPFall) to identify FRIDs when performing medica-
tion review [51]. As described earlier, studies have shown that the effect of FRIDs
on fall risk appears to be dependent on characteristics [49]. Thus, these characteris-
tics, including polypharmacy, frailty status, comorbidities, patient’s preferences,
and other geriatric syndromes, should be taken into account when performing a
medication review [65]. To succeed in this, the medication review is optimally
embedded in a holistic assessment to enable personalized medication strategy.
Furthermore, it is important to realize that in practice, when deprescribing of FRIDs
is initiated, compliance is notoriously poor, especially for psychotropic medications
168 L. Seppala and N. van der Velde
[66]. Thus, a single one-time advice does not suffice. For the long-term success of
deprescribing provision of support, monitoring, and documentation are essential
[67]. Monitoring should be arranged on individual basis [24]. Finally, about 10–15%
of falls result in fractures, and therefore, in the medication review, the known drugs
to affect skeletal fragility should also be considered [20].
The generic steps of medication management of older fallers are shown in
Fig. 13.1 [20]. Further practical guidance regarding deprescribing of FRIDs is
provided in the STOPPFall deprescribing tool [24]. The tool is a freely available
online decision support tool which works in a stepwise manner containing all the
steps of the decision tree of the Fig. 13.1 separately for each STOPPFall medication
class [68]. For example, for antidepressants, it states that at least in the following
cases deprescribing should be considered: (1) hyponatremia, orthostatic hypoten-
sion, dizziness, sedative symptoms, or tachycardia/arrhythmia; (2) if given for sleep
disorder; and (3) if given for depression, but this is dependent on symptom-free time
and history of symptoms [24]. Furthermore, the tool indicates that stepwise depre-
scribing is needed and after deprescribing at least the following symptoms should
be monitored: recurrence of depression, anxiety, irritability, and insomnia [24]. Also
a paper version is available in the Appendix of the publication [24].
13.8 Conclusion
Falls have a major impact on both individual and societal level. Mostly falls tend to
occur due to several interacting risk factors in an individual. Some of the key risk
factors are older age, past falls, mobility, and certain medication use (fall risk-
increasing drugs). Several psychotropic medications (benzodiazepines and related
drugs, antidepressants, antipsychotics), anticholinergics (such as overactive bladder
and incontinence medications and antihistamines), opioids, alpha-blockers, and
other cardiovascular medications (diuretics and centrally acting antihypertensives)
are considered to induce increased fall risk. FRIDs have cardiovascular, motoric,
and central nervous system related adverse effects which can lead to falls. Medication
review with the aim of FRID deprescribing as a part of multifactorial fall prevention
strategy is effective in fall prevention. This medication review should be a holistic
assessment including the evaluation of characteristics such as frailty and patient
goals to produce a personalized medication optimization plan. Regular medication
reviews will help to keep the exposure to FRIDs as short as possible. Structured
tools (such as STOPPFall) can aid with FRID deprescribing.
References
1. Berry SD, Miller RR. Falls: epidemiology, pathophysiology, and relationship to fracture. Curr
Osteoporos Rep. 2008;6(4):149–54.
2. Montero-Odasso M, van der Velde N, Alexander NB, Becker C, Blain H, Camicioli R, et al.
New horizons in falls prevention and management for older adults: a global initiative. Age
Ageing. 2021;50(5):1499–507.
3. Morris R, O’Riordan S. Prevention of falls in hospital. Clin Med (Lond). 2017;17(4):360–2.
4. Bouldin EL, Andresen EM, Dunton NE, Simon M, Waters TM, Liu M, et al. Falls among adult
patients hospitalized in the United States: prevalence and trends. J Patient Saf. 2013;9(1):13–7.
5. Step safely: strategies for preventing and managing falls across the life-course. Geneva: World
Health Organization; 2021. Licence: CC BY-NC-SA 3.0 IGO.
6. Milat AJ, Watson WL, Monger C, Barr M, Giffin M, Reid M. Prevalence, circumstances and
consequences of falls among community-dwelling older people: results of the 2009 NSW falls
prevention baseline survey. N S W Public Health Bull. 2011;22(3–4):43–8.
7. Heinrich S, Rapp K, Rissmann U, Becker C, König H-H. Cost of falls in old age: a systematic
review. Osteoporos Int. 2010;21(6):891–902.
170 L. Seppala and N. van der Velde
8. Ganz DA, Latham NK. Prevention of falls in community-dwelling older adults. N Engl J Med.
2020;382(8):734–43.
9. Deandrea S, Lucenteforte E, Bravi F, Foschi R, La Vecchia C, Negri E. Risk factors for falls
in community-dwelling older people: a systematic review and meta-analysis. Epidemiology.
2010;21(5):658–68.
10. Kyrdalen IL, Thingstad P, Sandvik L, Ormstad H. Associations between gait speed and
well-known fall risk factors among community-dwelling older adults. Physiother Res Int.
2019;24(1):e1743.
11. Verghese J, LeValley A, Hall CB, Katz MJ, Ambrose AF, Lipton RB. Epidemiology of gait
disorders in community-residing older adults. J Am Geriatr Soc. 2006;54(2):255–61.
12. Pirker W, Katzenschlager R. Gait disorders in adults and the elderly : a clinical guide. Wien
Klin Wochenschr. 2017;129(3–4):81–95.
13. Osman A, Kamkar N, Speechley M, Ali S, Montero-Odasso M. Fall risk-increasing drugs and
gait performance in community-dwelling older adults: a systematic review. Ageing Res Rev.
2022;77:101599.
14. Nnodim JO, Yung RL. Balance and its clinical assessment in older adults—A review. J Geriatr
Med Gerontol. 2015;1(1):003.
15. Moreland JD, Richardson JA, Goldsmith CH, Clase CM. Muscle weakness and falls in older
adults: a systematic review and meta-analysis. J Am Geriatr Soc. 2004;52(7):1121–9.
16. Seene T, Kaasik P. Muscle weakness in the elderly: role of sarcopenia, dynapenia, and pos-
sibilities for rehabilitation. Eur Rev Aging Phys Act. 2012;9(2):109–17.
17. Hart LA, Phelan EA, Yi JY, Marcum ZA, Gray SL. Use of fall risk-increasing drugs around a
fall-related injury in older adults: a systematic review. J Am Geriatr Soc. 2020;68(6):1334–43.
18. Stam H, Harting T, van der Sluijs M, van Marum R, van der Horst H, van der Wouden JC,
et al. Usual care and management of fall risk increasing drugs in older dizzy patients in Dutch
general practice. Scand J Prim Health Care. 2016;34(2):164–70.
19. Shaver AL, Clark CM, Hejna M, Feuerstein S, Wahler RG Jr, Jacobs DM. Trends in fall-
related mortality and fall risk increasing drugs among older individuals in the United
States,1999–2017. Pharmacoepidemiol Drug Saf. 2021;30(8):1049–56.
20. Seppala LJ, van der Velde N, Masud T, Blain H, Petrovic M, van der Cammen TJ, et al. EuGMS
Task and Finish group on Fall-Risk-Increasing Drugs (FRIDs): position on knowledge dis-
semination, management, and future research. Drugs Aging. 2019;36(4):299–307.
21. de Vries M, Seppala LJ, Daams JG, van de Glind EMM, Masud T, van der Velde N. Fall-risk-
increasing drugs: a systematic review and meta-analysis: I. Cardiovascular drugs. J Am Med
Dir Assoc. 2018;19(4):371.e1–9.
22. Seppala LJ, van de Glind EMM, Daams JG, Ploegmakers KJ, de Vries M, Wermelink A, et al.
Fall-risk-increasing drugs: a systematic review and meta-analysis: III. Others. J Am Med Dir
Assoc. 2018;19(4):372.e1–8.
23. Seppala LJ, Wermelink A, de Vries M, Ploegmakers KJ, van de Glind EMM, Daams JG, et al.
Fall-risk-increasing drugs: a systematic review and meta-analysis: II. Psychotropics. J Am
Med Dir Assoc. 2018;19(4):371.e11–7.
24. Seppala LJ, Petrovic M, Ryg J, Bahat G, Topinkova E, Szczerbińska K, et al. STOPPFall
(Screening Tool of Older Persons Prescriptions in older adults with high fall risk): a Delphi
study by the EuGMS Task and Finish Group on Fall-Risk-Increasing Drugs. Age Ageing.
2020;50:1189.
25. Rivasi G, Kenny RA, Ungar A, Romero-Ortuno R. Effects of benzodiazepines on orthostatic
blood pressure in older people. Eur J Intern Med. 2020;72:73–8.
26. Capiau A, Huys L, van Poelgeest E, van der Velde N, Petrovic M, Somers A. Therapeutic
dilemmas with benzodiazepines and Z-drugs: insomnia and anxiety disorders versus increased
fall risk—a clinical review. Eur Geriatr Med. 2022.
27. Stroup TS, Gray N. Management of common adverse effects of antipsychotic medications.
World Psychiatry. 2018;17(3):341–56.
13 Falls and Impaired Mobility 171
14.1 Introduction
G. Bellelli (*)
School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
IRCCS San Gerardo Hospital Monza, Acute Geriatrics Unit, Monza, Italy
e-mail: [email protected]
A. Morandi
REFiT Barcelona Research Group, Parc Sanitari Pere Virgili and Vall d’Hebron Institut de
Recerca (VHIR), Barcelona, Spain
delirium development, and the prevention and treatment of delirium using the phar-
macological approaches in older patients outside of intensive care settings.
There is general agreement that delirium represents an altered reaction of the central
nervous system (CNS) to the effect of one or more precipitating factors that inter-
play with one or more predisposing factors of delirium [12]. Drugs, in this context,
may represent either a precipitating or a predisposing factor (or both).
The exact mechanisms of drug-induced delirium are yet to be completely under-
stood. However, neurotransmitter imbalances involving acetylcholine, dopamine,
and gamma aminobutyric acid (GABA) traversing the cortical and subcortical ner-
vous system pathways have been observed in drug-induced delirium [13]. It is
hypothesized that delirium may develop as the result of an alteration of function of
the ascending arousal system directly provoked by an effect of GABAergic seda-
tives, anesthetics, and antihistamine drugs [14]. GABA acting at GABA-A recep-
tors inhibits the release of dopamine GABA antagonist [15], or a sudden withdrawal
from a GABA agonist may increase the risk of a hyperdopaminergic state, which in
turn facilitates the action of glutamate at N-methyl-D-aspartate (NMDA) receptors
[16]. This can explain why an abrupt benzodiazepine withdrawal may precipitate
delirium. In addition, this can explain why an imbalance of the brain cholinergic
and dopaminergic activity, which can be induced by a broad list of drugs with mus-
carinic antagonist activity, is also involved in delirium onset [14, 17–19]. Indeed,
drugs acting on GABA and NMDA receptors or with dopaminergic activity, such as
digoxin, quinolone antibiotics, morphine, and histamine (H2) receptor blockers, are
well-known causes of delirium [20].
Importantly, the receptor distribution of cholinergic and dopaminergic neu-
rotransmitters among layers of prefrontal cortex and temporal lobe indirectly sug-
gests that neurons of these areas could interact with each other during delirium [18].
It follows that a drug prescribed, for instance, to calm an agitated patient may lead
to a completely different effect in specific circumstances and patients, depending on
the receptor subtype which is more prevalent in those cortical areas. Older adults are
particularly sensitive to anticholinergic medication adverse effects because of
increased permeability of the blood-brain barrier, heightened sensitivity of cholin-
ergic receptors, deficient drug metabolism/elimination, and deficits in central cho-
linergic transmission. Anticholinergic activity has also been associated with an
increased severity of delirium [21].
The incidence of drug-induced delirium is particularly high even among older
individuals with multimorbidity and polypharmacy. Multimorbidity refers to a
coexistence of two or more chronic conditions in the same individual, and polyphar-
macy is commonly defined as the chronic use of five or more prescribed drugs [22,
23]. Patients with multimorbidity have an increased odd of assuming multiple medi-
cations and are thus at risk of polypharmacy prescription [24]. In addition, they
commonly have other co-occurring conditions, such as malnutrition, muscle mass
14 Optimizing Pharmacotherapy in Older Patients: Delirium 175
reduction, and low albumin serum levels, which leads to alterations in distribution,
with water-soluble drugs having a reduced volume of distribution and lipophilic
drugs having increased volume of distribution [25].
Aloisi and colleagues evaluated the association between polypharmacy and
delirium in a cohort of 4133 older inpatients, of which 23.4% had delirium [26]. The
authors found that polypharmacy was higher in patients with delirium, but the asso-
ciation was statistically significant only in surgical but not medical units. A possible
explanation was that in surgical settings, where the healthcare professionals were
not trained in geriatric care, older individuals were relatively overtreated with psy-
choactive drugs. Indeed, patients with delirium in these units had higher prescrip-
tion of benzodiazepines and antipsychotics [26]. However, another study including
410 consecutive patients admitted to an acute geriatric ward, of whom 25% devel-
oped delirium, found that polypharmacy was an independent risk factor for delir-
ium, after adjusting for covariates [27].
In addition to polypharmacy, Catic identified the following risk factors that
placed elders at increased risk of delirium: >9 chronic medications, >12 doses of
medication per day, >6 concurrent chronic diagnoses, prior adverse drug reaction,
low body weight, estimated creatinine clearance <50 mL/min, and age
>85 years [20].
Overall, the systematic review found that ARS was the only scale which was
consistently associated with delirium and thus supports its use. On the contrary,
when ADB was assessed with other scales, the findings were inconclusive.
However, delirium can be precipitated also by drugs which have not relevant
anticholinergic properties, such as opioids, steroids, and sedatives [4]. Therefore,
other researchers have developed other scales to assess a patient’s drug burden of
delirium. The Delirium Drug Scale (DDS) is a tool evaluation scale developed to
this aim [40, 41]. A rank of 1 or 2 is attributed to each member of the DDS drug list,
and to calculate the DDS score, the patient’s medication list is compared to the
DDS. The rank attributed to the drug is multiplied by the dosage factor to give a
weighted rank. The dosage factor is the result of the administered daily dose taken
divided by the sum of the administered daily dose and the minimal daily geriatric
dose (as reported by the Geriatric Dosage Handbook). The DDS score is the sum of
all weighted rank present in the patient’s drug list [40]. In an observational retro-
spective study on 1205 older individuals, Nguyen and colleagues found that the
DDS was associated with an increased risk of delirium incidence [41]. Importantly,
the risk of delirium increased as the DDS score rises from low to high exposure,
independently of risk and precipitating factors for delirium. The authors concluded
that the DDS could act as a decision tool for physicians when a new medication is
added to a patient’s drug therapy [41]. However, further research is required to con-
firm their results.
In addition, it is also important to highlight those other causal mechanisms, unre-
lated to drugs, can interfere with neurotransmission. For instance, during a sepsis, a
brain neuroinflammatory response triggered by inflammatory cytokines is com-
monly seen, leading to endothelial damage, disruption of the blood-brain barrier,
impaired blood flow, and neuronal apoptosis. Neuroinflammation can lead to
microglial overactivation, resulting in a neurotoxic response with further neuronal
injury. In these conditions, weighting the contribution of a specific drug to delirium
development might not always be possible, since the different inflammatory factors
and neurotransmitters are closely intertwined [42].
The American Geriatrics Society (AGS) Beers Criteria are an explicit list of poten-
tially inappropriate medications that, under specific situations, should be avoided in
older adults. The AGS Beers Criteria are updated on a 3-year cycle by an interdisci-
plinary expert panel that periodically reviews the evidence published since the last
update and then releases their recommendations [43, 44]. In the last update (i.e.,
2019), the list of drugs that should be avoided in people with delirium includes the
H2-receptor antagonists, the benzodiazepine (short, intermediate, and long acting),
the benzodiazepine receptor agonist hypnotics (i.e., the “Z drugs”: zolpidem, eszop-
iclone, and zaleplon), first (conventional) and second (atypical) generation antipsy-
chotics, and some pain medications, such as meperidine. In addition, the AGS Beers
14 Optimizing Pharmacotherapy in Older Patients: Delirium 177
Criteria recommend that all drugs acting on the central nervous system should be
avoided in older adults at high risk of delirium because of their potential to precipi-
tate delirium. In general, it is recommended that antipsychotics should not be pre-
scribed for behavioral problems of dementia and/or delirium unless
nonpharmacological options have failed or are not possible and the older adult is
threatening substantial harm to self or others [45].
The STOPP/START (Screening Tool of Older People’s Prescriptions and
Screening Tool to Alert to Right Treatment) criteria include a list of both potential
inappropriate medications and potential prescribing omissions in older people [46,
47], which was developed by a panel of European experts. There are not explicit
recommendations to avoid the use of medications for preventing delirium; however,
it is recommended to avoid tricyclic antidepressants in individuals with dementia,
the long-term use of long-acting benzodiazepines (e.g., chlordiazepoxide, fluraze-
pam, nitrazepam, chlorazepate, and diazepam), and the long-term use of neurolep-
tics as hypnotics, especially in those with parkinsonism, as well as anticholinergics
to treat extrapyramidal side effects of neuroleptic medications.
In 1999, Inouye and colleagues proposed firstly the Hospital Elder Life Program
(HELP), a model of care tailored for older patients and specifically oriented to pre-
vent delirium during the hospital stay, including cognitive stimulation, sleep
enhancement, promotion of mobility, and avoidance of dehydration and sensory
impairments (i.e., visual and hearing impairments) [48]. Since then, several studies,
systematic reviews, and meta-analysis have confirmed the effectiveness of non-
pharmacological, multicomponent, and multidisciplinary interventions in delirium
prevention [49, 50]. The most recently updated delirium guidelines support such
approach as the first step for delirium prevention [51], leading to a reduction of
delirium incidence by almost 43%.
On the contrary, there is insufficient evidence to recommend the use of specific
medications for delirium prevention [52]. Fok and colleagues reported a possible
role on the use of antipsychotics to reduce the incidence of postoperative delirium,
mainly in orthopedic settings and in those patients at higher risk of delirium [53].
However, a subsequent systematic review investigating the possible effect of halo-
peridol vs. placebo in adult patients reported a limited evidence of a small dose of
haloperidol in delirium reduction among surgical patients [54]. Therefore, guide-
lines suggest that the pharmacological risk reduction should occur only by acting on
three specific areas of intervention: (a) medication reconciliation with a thorough
review of medication including over-the-counter and herbal medications; (b) the
physiological changes related to aging and their effects on medication metabolism;
and (c) considering the risk of delirium related to the introduction of a new drug [52].
Recent studies have claimed potential applications for some medications. For
example, there is emerging evidence that dexmedetomidine—a highly selective
α2-adrenergic agonist—alone or in combination with other sedatives may reduce
178 G. Bellelli and A. Morandi
postoperative delirium [55]. This type of medication has been widely studied in
critically ill patients admitted to intensive care units (ICU) [56], and its potential use
outside the ICU needs to be further investigated.
Given that sleep-cycle alterations are often present in patients with delirium and
especially in delirium superimposed on dementia, there has been an increasing interest
on the effect of melatonin and melatonin receptor agonist (i.e., ramelteon) for delirium
prevention. A recent meta-analysis showed that both melatonin and ramelteon are
effective in reducing delirium incidence in hospitalized patients, but this effect was
found significant in surgical patients and not in medical patients [57]. Furthermore, in
a recent multicenter randomized placebo-controlled trial, Hatta et al. showed that
suvorexant (an orexin inhibitor used in the treatment of insomnia) significantly reduced
the occurrence of delirium compared to the administration of placebo [58]. A meta-
analysis that included seven studies conducted in patients undergoing this treatment
compared with controls showed that delirium incidence could be reduced in the treat-
ment groups compared to controls [59]. However, further larger studies are required.
14.7 Conclusions
This is a very important area of research that promises to provide exciting news
for clinicians and opportunities for the patients.
References
1. American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th
ed. Arlington: American Psychiatric Association; 2013.
2. Bellelli G, Morandi A, Di Santo SG, et al. “Delirium Day”: a nationwide point prevalence
study of delirium in older hospitalized patients using an easy standardized diagnostic tool.
BMC Med. 2016;14(1):106. https://doi.org/10.1186/s12916-016-0649-8.
3. Morandi A, Di Santo SG, Zambon A, et al. Delirium, dementia and in-hospital mortality: the
results from the Italian Delirium Day 2016, a national multicenter study. J Gerontol A Biol Sci
Med Sci. 2019, 74(6):910–6. https://doi.org/10.1093/gerona/gly154.
4. Inouye SK, Westendorp RG, Saczynski JS. Delirium in elderly people. Lancet.
2014;383(9920):911–22. https://doi.org/10.1016/S0140-6736(13)60688-1.
5. Morandi A, Davis D, Fick D, et al. Delirium superimposed on dementia strongly predicts
worse outcomes in older rehabilitation inpatients. J Am Med Dir Assoc. 2014;15(5):349–54.
https://doi.org/10.1016/j.jamda.2013.12.084.
6. Wilcox ME, Girard TD, Hough CL. Delirium and long term cognition in critically ill patients.
BMJ. 2021;373:n1007. https://doi.org/10.1136/bmj.n1007.
7. Pereira JV, Aung Thein MZ, Nitchingham A, Caplan GA. Delirium in older adults is associ-
ated with development of new dementia: a systematic review and meta-analysis. Int J Geriatr
Psychiatry. 2021;36(7):993–1003. https://doi.org/10.1002/gps.5508.
8. Morandi A, Lucchi E, Turco R, et al. Delirium superimposed on dementia: a quantitative and
qualitative evaluation of informal caregivers and health care staff experience. J Psychosom
Res. 2015;79(4):272–80. https://doi.org/10.1016/j.jpsychores.2015.06.012.
9. Morandi A, Lucchi E, Turco R, et al. Delirium superimposed on dementia: a quantitative and
qualitative evaluation of patient experience. J Psychosom Res. 2015;79(4):281–7. https://doi.
org/10.1016/j.jpsychores.2015.07.010.
10. Leslie DL, Marcantonio ER, Zhang Y, Leo-Summers L, Inouye SK. One-year health care
costs associated with delirium in the elderly population. Arch Intern Med. 2008;168(1):27–32.
https://doi.org/10.1001/archinternmed.2007.4.
11. Gould J, Lond BS. Treatment of delirium, psychosis, and coma due to drugs. Lancet.
1953;1(6760):570–3. https://doi.org/10.1016/s0140-6736(53)91696-1.
12. Inouye SK, Charpentier PA. Precipitating factors for delirium in hospitalized elderly persons.
Predictive model and interrelationship with baseline vulnerability. JAMA. 1996;275(11):852–7.
13. Alagiakrishnan K, Wiens CA. An approach to drug induced delirium in the elderly. Postgrad
Med J. 2004;80(945):388–93. https://doi.org/10.1136/pgmj.2003.017236.
14. Wilson JE, Mart MF, Cunningham C, et al. Delirium. Nat Rev Dis Primers. 2020;6(1):90.
https://doi.org/10.1038/s41572-020-00223-4.
15. Hauber W. Involvement of basal ganglia transmitter systems in movement initiation. Prog
Neurobiol. 1998;56(5):507–40. https://doi.org/10.1016/s0301-0082(98)00041-0.
16. Cepeda C, Levine MS. Dopamine and N-methyl-D-aspartate receptor interactions in the neo-
striatum. Dev Neurosci. 1998;20(1):1–18. https://doi.org/10.1159/000017294.
17. Trzepacz PT. The neuropathogenesis of delirium. A need to focus our research. Psychosomatics.
1994;35(4):374–91. https://doi.org/10.1016/S0033-3182(94)71759-X.
18. Trzepacz PT. Is there a final common neural pathway in delirium? Focus on acetylcholine
and dopamine. Semin Clin Neuropsychiatry 2000;5(2):132–148. https://doi.org/10.153/
SCNP00500132.
19. Pasina L, Colzani L, Cortesi L, et al. Relation between delirium and anticholinergic drug
burden in a cohort of hospitalized older patients: an observational study. Drugs Aging.
2018;36:85–91. https://doi.org/10.1007/s40266-018-0612-9.
14 Optimizing Pharmacotherapy in Older Patients: Delirium 181
56. Burry LD, Cheng W, Williamson DR, et al. Pharmacological and non-pharmacological
interventions to prevent delirium in critically ill patients: a systematic review and net-
work meta- analysis. Intensive Care Med. 2021;47(9):943–60. https://doi.org/10.1007/
s00134-021-06490-3.
57. Khaing K, Nair BR. Melatonin for delirium prevention in hospitalized patients: a system-
atic review and meta-analysis. J Psychiatr Res. 2021;133:181–90. https://doi.org/10.1016/j.
jpsychires.2020.12.020.
58. Hatta K, Kishi Y, Wada K, et al. Preventive effects of Suvorexant on delirium: a random-
ized placebo-controlled trial. J Clin Psychiatry. 2017;78(8):e970–9. https://doi.org/10.4088/
JCP.16m11194.
59. Xu S, Cui Y, Shen J, Wang P. Suvorexant for the prevention of delirium: a meta-analysis.
Medicine (Baltimore). 2020;99(30):e21043. https://doi.org/10.1097/MD.0000000000021043.
60. Flaherty JH, Tariq SH, Raghavan S, Bakshi S, Moinuddin A, Morley JE. A model for man-
aging delirious older inpatients. Research Support, U.S. Gov’t, P.H.S. J Am Geriatr Soc.
2003;51(7):1031–5.
61. Neufeld KJ, Yue J, Robinson TN, Inouye SK, Needham DM. Antipsychotic medication for
prevention and treatment of delirium in hospitalized adults: a systematic review and meta-
analysis. Meta-Analysis Research Support, N.I.H., Extramural Research Support, Non- -
U.S. Gov’t Review. J Am Geriatr Soc. 2016;64(4):705–14. https://doi.org/10.1111/jgs.14076.
62. Burry L, Mehta S, Perreault MM, et al. Antipsychotics for treatment of delirium in hospi-
talised non-ICU patients. Cochrane Database Syst Rev. 2018;6:CD005594. https://doi.
org/10.1002/14651858.CD005594.pub3.
63. Nikooie R, Neufeld KJ, Oh ES, et al. Antipsychotics for treating delirium in hospitalized
adults: a systematic review. Ann Intern Med. 2019;171(7):485–95. https://doi.org/10.7326/
M19-1860.
64. Barbateskovic M, Krauss SR, Collet MO, et al. Haloperidol for the treatment of delirium in
critically ill patients: a systematic review with meta-analysis and trial sequential analysis. Acta
Anaesthesiol Scand. 2020;64(2):254–66. https://doi.org/10.1111/aas.13501.
65. Agar MR, Lawlor PG, Quinn S, et al. Efficacy of oral risperidone, haloperidol, or placebo for
symptoms of delirium among patients in palliative care: a randomized clinical trial. JAMA
Intern Med. 2017;177(1):34–42. https://doi.org/10.1001/jamainternmed.2016.7491.
66. Hui D, Frisbee-Hume S, Wilson A, et al. Effect of lorazepam with haloperidol vs haloperidol
alone on agitated delirium in patients with advanced cancer receiving palliative care: a random-
ized clinical trial. JAMA. 2017;318(11):1047–56. https://doi.org/10.1001/jama.2017.11468.
67. Tampi RR, Tampi DJ, Ghori AK. Acetylcholinesterase inhibitors for delirium in
older adults. Am J Alzheimers Dis Other Dement. 2016;31(4):305–10. https://doi.
org/10.1177/1533317515619034.
68. van Eijk MM, Roes KC, Honing ML, et al. Effect of rivastigmine as an adjunct to usual care
with haloperidol on duration of delirium and mortality in critically ill patients: a multicentre,
double-blind, placebo-controlled randomised trial. Lancet. 2010;376(9755):1829–37. https://
doi.org/10.1016/S0140-6736(10)61855-7.
69. Li Y, Ma J, Jin Y, et al. Benzodiazepines for treatment of patients with delirium exclud-
ing those who are cared for in an intensive care unit. Cochrane Database Syst Rev.
2020;2:CD012670. https://doi.org/10.1002/14651858.CD012670.pub2.
Optimizing Pharmacotherapy in Older
Adults: Urinary Incontinence 15
Antoine Vella and Claudio Pedone
15.1 Introduction
Urinary incontinence (UI) in older persons is common and increases with age,
affecting around 70% of older persons over 85 years of age. It can have a major
adverse impact on their quality of life and is one of the reasons why older persons
end up in long-term care. Unfortunately, it is frequently underdiagnosed and often
not treated even though it is potentially reversible.
Management of patients suffering from UI requires not only a focused history,
examination, and investigations for an accurate diagnosis to be made but also a
comprehensive geriatric assessment that usually requires the involvement of a mul-
tidisciplinary team. Treatment options usually include lifestyle changes, behavioral
interventions, often pharmacotherapy, and less commonly surgical interventions
besides the use of appropriate devices and absorbent aids.
Pharmacotherapy in the incontinence setting must have a two-pronged approach.
Before considering specific pharmacotherapy targeting the incontinence, a compre-
hensive review of all ongoing medications, including those over-the-counter, must
be undertaken. This is because there are several drugs that, directly or indirectly, can
A. Vella (*)
Department of Geriatric Medicine, St. Vincent De Paul Hospital, Luqa, Malta
e-mail: [email protected]
C. Pedone
Department of Medicine and Surgery, Research Unit of Geriatrics, Università Campus
Bio-Medico di Roma, Rome, Italy
Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
e-mail: [email protected]
impact negatively on the patient’s continence status. This shall be discussed sepa-
rately below. The second aspect of the pharmacotherapeutic management can then
be directed more specifically at the patient’s continence problem.
Pharmacotherapy for incontinence should form part of a holistic approach and
must take the patients’ comorbidities, desires, and quality of life into account.
Treatment must be carefully monitored with special vigilance for side effects of
medications. The value of team management, preferably with a continence outreach
service, cannot be overstressed.
The mode of actions and effects that medications have on micturition, whether
they are medications that the patient is on or pharmacotherapy for incontinence, can
only be understood when the basis of the complex micturition process is
comprehended.
The lower urinary tract consists essentially of two units: the bladder which acts
as a reservoir and the outlet unit which consists of the bladder neck, the urethra, and
the external urethral sphincter (EUS). The bladder and bladder neck, including the
internal urethral sphincter (IUS), consist of smooth muscle, while the EUS consists
of striated muscle. The bladder has two modes of action—storage and elimina-
tion—and works in a switch on/off pattern which is under voluntary control. The
mechanisms that underlie the control of micturition consist of complex neural path-
ways that function at the levels of the brain, spinal cord, and peripheral nerves that
are mediated via multiple neurotransmitters at all levels. A complex description of
this is beyond the scope of the chapter, but a very simplified version of the efferent
neural pathways and neurotransmitter mechanisms that regulate the lower urinary
tract (LUT) will be summarized in Fig. 15.1.
The LUT has three main nerve supplies: the sympathetic, parasympathetic, and
somatic systems [1].
The sympathetic system via the hypogastric nerve is inhibitory to the detrusor
muscle and motor to the IUS. Sympathetic postganglionic neurons release nor-
adrenaline (NA) which activates the following:
L3
NO (-) NA (+)
r EUS contraction S4
a
Fig. 15.1 Lower urinary tract: efferent innervation and neurotransmitter–receptor mechanisms
15 Optimizing Pharmacotherapy in Older Adults: Urinary Incontinence 187
The parasympathetic system via the pelvic nerve plexus is motor to the detrusor
muscle and inhibitory to the IUS. When the bladder fills to beyond 300 mL, stretch
mechanoreceptors in the bladder wall relay via pelvic splanchnic nerves to the spi-
nal cord. Efferent parasympathetic nerves synapse with postganglionic nerves in the
bladder wall and IUS via acetylcholine (ACh) acting on muscarinic receptors (M).
This results in the following:
–– Bladder contraction
–– Relaxation of the IUS
The somatic system via the pudendal nerve is motor to the EUS which is under
voluntary control and is always contracted except during micturition when the EUS
is inhibited. The somatic axons release acetylcholine which via nicotinic receptors
(NR) result in the following:
–– EUS contraction
Antimuscarinic
(Bladder relaxation)
b receptor agonist
Parasympathetic Pelvic Nerves (Bladder relaxation) Sympathetic (Hypogastric) Nerve
U a1 a receptor blockers
IUS (IUS relaxation) Somatic (Pudendal) Nerve
BoNT/A r
Bladder relaxation e
S2
t Ach (+)
EUS h NR S3
r duloxetine S4
a (increase in EUS tone during
storage phase of micturition)
Fig. 15.2 Mechanisms of action of therapeutic agents on the lower urinary tract
188 A. Vella and C. Pedone
pontine center synchronizes and maintains sustained bladder contractions, and the
cortical center inhibits the pontine center until a suitable time and place for micturi-
tion is available. These centers are regulated by several neurotransmitters which can
be affected by disease processes a well as medications that the patients are taking.
rate from treatment. Adverse effects can be short term, e.g., nausea with duloxetine for
SUI, but can also be long term, e.g., worsening of cognitive impairment with antimus-
carinic agents in predisposed older patients. Treatment for BOO is usually better toler-
ated, but long-term adverse effects such as erectile dysfunction can be a problem.
One must therefore carefully weigh the risk of adverse effects, taking into con-
sideration the particular patient’s comorbidities, quality of life, and wishes, before
embarking on treatment. Evidence of efficacy of these medications in the frail older
person is scant, and patients that started on these medications should be monitored
vigilantly for adverse effects.
Antimuscarinic drugs are the mainstay of the drug therapy for overactive bladder
(OAB), with or without urinary urge incontinence (UUI). These drugs seem to be
effective in the short term, while data on long-term efficacy are scant [3]. Overall,
the effect on incontinence itself is limited (about 20% reduction of incontinence
episodes), but the effect on quality of life seems to be more substantial. Evidence
pertaining to efficacy in older and frail people is limited. Side effects due to sys-
temic anticholinergic action are common and frequently lead to therapy discontinu-
ation. In older people, negative effects on cognition are of some concern, and these
drugs should be avoided in patients with cognitive impairment.
15.5.1.2 Evidence
In older people, antimuscarinic drugs improve the severity of urinary incontinence,
although the effect size is generally small and no data are available on their long-
term efficacy. No evidence is available, however, in older people with multiple mor-
bidities and polypharmacotherapy. Side effects are common and related to systemic
anticholinergic activity: dry mouth, constipation, blurred vision, and dizziness.
These side effects have a sizable impact on persistence on treatment: about 40% of
patients discontinue these drugs within 3 months of therapy.
β3 receptor agonists (mirabegron and vibegron) are generally considered safe, but
data specific for older people is scant. In particular, their effect on the QT interval in
people also taking drugs known to cause QT prolongation is unknown.
15.5.2.2 Evidence
A meta-analysis of head-to-head studies has shown that mirabegron is as effective
as solifenacin in reducing the burden of OAB/UUI, with comparable rates of adverse
events [4].
cytochrome. Furthermore, they should not be used in patients with poorly controlled
arterial hypertension.
Botulinum toxin A (BoNT/A) could be offered to patients with UUI who are refrac-
tory to treatment with antimuscarinics or beta 3 receptor agonists. This therapy is
not without side effects, and patients need to be counseled before embarking on this
treatment. Evidence in frail older persons is limited.
15.5.3.2 Evidence
BoNT/A infiltration for UUI has been shown to decrease the number of micturitions
and episodes of UI per day and increase bladder filling capacity and the volume of
urine per voiding episode. In a meta-analysis comparing BoNT/A treatment to pla-
cebo, 30% of the patients were cured against 7% in the placebo arm. Quality of life
at 6 months was higher in the treatment group. A study comparing BoNT/A to anti-
muscarinics showed similar rates of improvement in the first 6 months, but the cure
rate was higher in the BoNT/A arm [6]. However, there were higher rates of urinary
tract infections (UTIs) and retention of urine than with the antimuscarinics. These
side effects as well as the need for this treatment to be repeated at 5–9-month inter-
vals result in a high dropout rate from treatment which has been shown to be 60%
at 5-year follow-up [7].
catheterization (CIC) as well as UTIs (30%). Risk factors for adverse effects
include age over 61 years as well as a low maximal flow rate, low voiding effi-
ciency, and a high PVR baseline prior to starting treatment. Evidence for use in the
frail older person is limited, and CIC in this patient group can prove difficult to
perform safely.
15.6.1 Duloxetine
15.6.1.2 Evidence
Duloxetine improves SUI, mixed urinary incontinence (MUI), and to a lesser extent
UUI in women. It can improve SUI in males and has also been shown to decrease
post-prostatectomy leakage [6].
It is more effective than PFMT alone in reducing leakage although a trial of
PFMT is usually recommended prior to commencing the medication. However,
PFMT works synergistically with duloxetine, and the effect of the latter on quality
of life (QoL) is better when combined with PFMT.
194 A. Vella and C. Pedone
15.6.2 Desmopressin
15.6.2.2 Evidence
Although it has been shown to be beneficial in the treatment of nocturia, there is no
evidence to support its use in UI. Two RCTs have shown some decrease in daytime
15 Optimizing Pharmacotherapy in Older Adults: Urinary Incontinence 195
incontinence in the 4-h period following its administration; however, this effect
was not sustained thereafter. Its long-term use does not improve UI in men
and women.
Estrogen receptors have been found in the pelvic floor muscles, urogenital liga-
ments, the epithelial tissue of the bladder, urethra and trigone, detrusor muscle,
and vagina.
Estrogens produce a trophic effect on the epithelial mucosa in the vagina, ure-
thra, and bladder in this way enhancing the “mucosal sphincter” effect as well as
increasing the periurethral vascularization which is important in regulating the ure-
thral closing pressure [12]. They also increase the ratio of alpha to beta adrenergic
receptors favoring increased urethral tonicity. Some of the perceived improvement
in lower urinary tract symptoms might be because of their estrogenic effect of
decreasing vaginal discomfort associated with vaginal atrophy.
The effects of estrogens on incontinence comes about from the fact that estrogen
receptors are ubiquitous in the LUT and therefore maintain a stable balance between
tissues that confers improvement in postmenopausal UI.
15.7.2 Evidence
In men, one of the most common causes of overactive bladder and urge incontinence is
benign prostatic hypertrophy (BPH). In people with this condition, the association of
drugs for BPH and antimuscarinic/mirabegron may be used, although the latter must be
avoided in the presence of severe obstruction. Among drugs used for BPH, both
5-alpha-reductase inhibitors (including Serenoa repens) and alpha-adrenergic antago-
nists are effective in reducing overactive bladder symptoms. In the older people, how-
ever, alpha-adrenergic antagonists are associated with increased risk of hypotension
and falls, and while the risk is smaller with prostate-specific drugs, it is still significant.
Serenoa repens is also used to treat symptoms. Phosphodiesterase inhibitors such as
tadalafil may also be used as adjuvant therapy in selected subset of patients.
15.8.2 Evidence
however, may be less tolerated by older patients because of the risk of orthostatic
hypotension. Evidence on the effect of Serenoa repens are conflicting.
15.9 Conclusions
References
1. Fowler CJ, Griffiths D, de Groat WC. The neural control of micturition. Nat Rev Neurosci.
2008;9:453–66.
2. Aharony L, De Cock J, Nuotio M, Pedone C, Vande Walle N, Velge A, Vella A, Verdejo
BC. Consensus document on the management of urinary incontinence in older persons. Eur
Geriatr Med. 2017;8:202–9.
3. Nabi G, Cody JD, Ellis G, Hay-Smith J, Herbison GP. Anticholinergic drugs versus placebo
for overactive bladder syndrome in adults. Cochrane Database Syst Rev. 2006;4:CD003781.
4. Wang J, Zhou J, Cui Y, Li Y, Yuan H, Gao Z, Zhu Z, Wu J. Meta-analysis of the efficacy and
safety of mirabegron and solifenacin monotherapy for overactive bladder. Neurourol Urodyn.
2019;38:22–30.
5. Hsieh PF, Hung-Chieh C, Chao-Hsiang C, Chieh-Lung CE. Botulinum toxin A for the treat-
ment of overactive bladder. Toxins. 2016;8:59. https://doi.org/10.3390/toxins8030059.
6. Lucas MG, Bedretdinova D, Berghmans LC, Bosch JLHR, Burhard FC, Cruz F et al.
Guidelines on urinary incontinence—partial update. 2015 European Association of Urology.
7. Mohee A, Khan A, Harris N, Eardley I. BJU Int. 2012;111:106–13. https://doi.
org/10.1111/j.1464-410x.2012.11282.x.
198 A. Vella and C. Pedone
8. Abreu-Mendes P, Silva J, Cruz F. Pharmacology of the lower urinary tract: update on LUTS
treatment. Ther Adv Urol. 2020;12:1–16. https://doi.org/10.1177/1756287220922425.
9. Schuessler B. What do we know about Duloxetine’s mode of action? BJOG. 2006;113(Suppl
1):5–9. https://doi.org/10.1111/j.1471-0528.2006.00877.x.
10. Jinhang L, Yang L, Chunxiao P, Tana Y, Haichao Y, Han P. The role of duloxetine in stress uri-
nary incontinence: a systematic review and meta-analysis. Int Urol Nephrol. 2013;45:679–86.
https://doi.org/10.1007/s11255-013-0410-6.
11. Weatherall M. The risk of hyponatremia in older adults using desmopressin for nocturia: a
systematic review and meta-analysis. Neurourol Urodyn. 2004;23:302–5.
12. Legendre G, Ringa V, Fauconnier A, Fritel X. Menopause, hormone treatment and urinary
incontinence at mid-life. Maturitas. 2013;74:26–30.
13. Weber MA, Lim V, Oryszcyn J, te West N, Soujet J, Jeffrey S, Roovers JP, Moore KH. The
effect of vaginal oestriol cream on subjective and objective symptoms of stress urinary incon-
tinence and vaginal atrophy: an international multi-centre pilot study. Gynecol Obstet Invest.
2017;82:15–21. https://doi.org/10.1159/000445074.
14. McConnell JD, Roehrborn CG, Bautista OM, Andriole GL Jr, Dixon CM, Kusek JW, Lepor
H, McVary KT, Nyberg LM Jr, Clarke HS, Crawford ED, Diokno A. The long-term effect of
doxazosin, finasteride, and combination therapy on the clinical progression of benign prostatic
hyperplasia. N Engl J Med. 2003;349:2387–98.
15. Takahashi S, Kato K, Takei M, Yokoyama O, Gotoh M. Efficacy and safety of the noradrenaline
reuptake inhibitor, TAS-303, in women with stress urinary incontinence: results of a double-
blind, randomized, placebo-controlled, early phase II trial. Int J Urol. 2021;28(1):82–90.
https://doi.org/10.1111/iju.14411.
Constipation
16
Giammarco Fava
16.1 Lifestyle
General measures such as increased fluid intake and exercise are suggested to man-
age constipation (Fig. 16.1). Even if poor evidences support this advice [9], it is
suggested increasing water intake to 1.5–2 L daily to enhance the effects of fiber in
constipated patients [10] (Fig. 16.1). Patients with a normal bowel transit usually
empty stools at approximately the same time daily [11, 12], and defecation is par-
tially a conditioned reflex. Colonic motor activity is more active after waking and
after a meal [13]; thus, the optimal time for defecation is usually within the first 2 h
after waking and after breakfast. For this reason, there is a rationale in educating
patients to attempt a bowel movement at least twice a day, usually 30 min after
meals, and to strain for no more than 5 min [14].
G. Fava (*)
Gastroenterology and Digestive Endoscopy Unit, Italian National Research Center on Aging
(IRCCS-INRCA), Ancona, Italy
e-mail: [email protected]
Constipation (primary)
no response response
continue
Osmotic laxatives
PAMORAs
In opioid-induced response
no response
constipation continue, adjusting dosage
Stimulant laxatives (temporary)
no response response
reducing dosage
Loosing
Colonic secretagogues
response
5HT4 receptor agonists
no response response
continue, adjusting dosage
Reassess diagnosis
Surgery
Fiber increases stool bulk, which causes colonic distention and promotes stool pro-
pulsion. Fiber is obtained primarily from grains and bran cereals [12]. There is a
weak evidence for the effectiveness of fiber consumption as treatment of constipa-
tion in older people. One study demonstrates that higher fiber intake was associated
with lower laxative use among older women [15], but another work shows that
higher intake of bran was associated with no reduction in constipation symptoms
and greater fecal loading in the colon [5]. In practical terms, a daily fiber intake of
20–30 g/day is generally recommended to improve bowel transit (Fig. 16.1). The
effects of fiber on bowel movements may take several weeks. Bloating, flatulence,
and cramps are a common problem with increased fiber intake, especially bran
fiber [16].
16 Constipation 201
Laxatives are prescribed in patients with constipation not responding to lifestyle and
dietary modification. Many studies comparing laxative effects in older people are of
poor quality and limited by unclear definitions for constipation, inconsistent out-
come measurement, and underreporting of potential confounding factors during the
trial period (e.g., fiber intake) [17–26]. Laxatives are used daily by 10–18% of com-
munity dwelling older adults and 74% of nursing home residents [4, 27, 28]. These
data suggest that sometimes these drugs are empirically prescribed to older patients
[29, 30]. However, osmotic salts, sugars and sugar alcohols, polyethylene glycol
(PEG), anthraquinones, diphenolic laxatives, and diphenylmethanes generally have
been shown efficacy and safety [17, 29, 30].
Osmotic laxatives should be used in patients not responding to bulk forming laxa-
tives [20] (Fig. 16.1). Osmotic laxatives increase fecal volume and reduce stool
consistency by creating an intraluminal osmotic gradient that leads to secretion of
202 G. Fava
water and electrolytes into the intestinal lumen [17]. They are divided into three
categories: poorly absorbed ions, nonabsorbable carbohydrates, and polyethyl-
ene glycol.
Magnesium, sulfate, and phosphate ions are partly absorbed by the intestine, thus
creating a hyperosmolar intraluminal environment. This leads to an osmotic mecha-
nism, even if they have other possible effects with unclear consequences, such as
increase of prostaglandin concentrations in the stool [21]. Magnesium hydroxide
typically induces a bowel movement within 6 h (Table 16.1). Moreover, magnesium
sulfate is a potent laxative that produces a large volume of liquid stool and often
leads to abdominal distention and sudden passage of a foul-smelling liquid stool.
However, the assumption of magnesium, particularly in older adults, needs to be
limited since it could cause adverse effects such as flatulence, abdominal cramps,
and volume overload. A small amount of magnesium is actively absorbed in the
small intestine, while the remainder draws water into the intestine by an osmotic
gradient [22]. Hypermagnesemia can develop particularly in patients with kidney
diseases and in children, and hypermagnesemia-induced paralytic ileus was
described as a rare complication [23]. Absorption of magnesium can cause electro-
lyte and volume overload in patients with kidney failure or cardiac dysfunction.
Also, in case of dehydration, excessive use of magnesium based laxatives can
develop these complications even in healthy patients.
Phosphate is mostly absorbed by the small intestine; thus, a large quantity needs
to be ingested to produce an osmotic laxative effect, and this is not ideal for routine
use [17]. Rare but serious reports of acute phosphate nephropathy have been
described in patients treated with oral sodium phosphate products for colon cleans-
ing prior to colonoscopy. Some patients have resulted in permanent damage of renal
function and required long-term dialysis. While some cases have occurred in
patients with normal baseline renal function, risk factors of acute phosphate
nephropathy include advanced age, hypovolemia, increased bowel transit time (such
as bowel obstruction), active colitis, kidney disease, and use of medicines that affect
renal perfusion or function such as diuretics, angiotensin-converting enzyme [ACE]
inhibitors, angiotensin receptor blockers, and possibly nonsteroidal anti-
inflammatory drugs [NSAIDs]. Patients should be advised to split dosage regimen
and warning about the importance of adequate hydration before, during, and after
use of oral sodium phosphate products. Additional sodium phosphate-based purga-
tive or enema products should be avoided [24].
Osmotic laxatives
Nonabsorbable Metabolically Stool
Poorly absorbed ions carbohydrates inert Stimulant laxatives Colonic secretagogues Serotoninergics softener Enemas Suppositories
Drugs (generic Bulking agents Magnesium Sodium Lactulose PEG Senna Bisacodyl Lubiprostone Linaclotide Prucalopride Docusate Phosphate enema Bisacodyl Bisacodyl Glycerin
name) (psyllium, hydroxide phosphate sodium
methylcellulose,
polycarbophil)
Recommended 4–20 g 15–30 mL once 20 mL once 15–30 mL 17–34 g once 5–20 mL 10–30 mg 24 μg twice 290 μg 2 mg once 100 mg 120 mL once 10 mg 10 mg 60 g
dose (daily) or twice once or twice or twice once; once once twice once once once
7.5–
15 mg
once
Side effects Bloating, Bloating, Acute Bloating, Bloating, Cramps, Cramps, Headache, Diarrhea, Nausea, Efficacy Rectal mucosa Rectal Rectal Rectal
intestinal abdominal phosphate diarrhea, cramps, diarrhea, nausea, nausea, bloating headache, not well injury, mucosa mucosa mucosa
obstruction, cramps, volume nephropathy nausea, flatulence, nausea, electrolyte diarrhea diarrhea defined hyperphosphatemia irritation irritation irritation
allergic overload (in pts. (during hyperglycemia electrolyte vomiting, imbalance,
reactions (rare) with kidney bowel in diabetics imbalance, melanosis vomiting
disease or cardiac preparation) allergic coli
dysfunction), reaction,
hypermagnesemia Mallory
Weiss tears
(rare, during
bowel prep)
203
204 G. Fava
the small intestine and is transformed in the colon to yield short-chain fatty acids,
hydrogen, and carbon dioxide, with consequent pulling water out from the body and
into the colon and lowering of the fecal pH. If lactulose is administered at dosage of
20 g (30 mL) daily, the sugar is not detectable in the stool. Indeed, in larger doses,
some drug passes through the colon unchanged; thus, it should be used with caution
in diabetic patients because of the risk of increasing glycemia. The recommended
dose of lactulose in adults is 15–30 mL once or twice daily. The time to onset of
action is longer than that of other osmotic laxatives since 2 or 3 days are required
for lactulose to achieve an effect. Some patients report that lactulose is effective
initially but then gradually loses its effect, possibly due to alteration in the intestinal
flora in response to the medication [25]. Side effects related to lactulose include
abdominal distention or discomfort, presumably as a result of colonic gas produc-
tion, diarrhea, and nausea. Cases of lactulose-induced megacolon have been also
reported [25].
The osmotic laxatives are consistently better than placebo for improving the
symptoms of constipation [26, 31]. Four studies compared osmotic laxative with
placebo, and three studies compared two osmotic laxatives. In two RCTs, lactulose
significantly increased the defecation frequency in respect with placebo and signifi-
cantly decreased the need for laxative use [32, 33]. Lactitol also significantly
increased the number of defecations, improved stool consistency, and decreased
laxative use compared with placebo [34].
Sorbitol and Mannitol. Sorbitol is widely used in the food industry as an artificial
sweetener but is rarely used in clinical practice. Ingestion of 5 g sorbitol causes a
rise in breath hydrogen, and 20 g produces diarrhea in about half of normal subjects
[35]. Sorbitol is as effective as lactulose and less expensive. A randomized double-
blind crossover trial of lactulose (20 g/day) and sorbitol (21 g/day) in ambulatory
older men with chronic constipation showed no difference between the two com-
pounds with regard to frequency or normality of bowel movements or patient pref-
erence [36]. The frequency of side effects was similar except for nausea, which was
more common with lactulose. Mannitol is another sugar alcohol that can be used as
a laxative. Like sorbitol, it is rarely used for clinical management of constipation.
include abdominal bloating, cramps, and flatulence [37, 38] (Table 16.1). In addi-
tion, the most commonly reported side effects of PEG used for colonoscopy prepa-
ration include electrolyte imbalances, allergic reactions, and Mallory-Weiss tears
[39] (Table 16.1). Cases of fulminant pulmonary edema have been reported after
administration of PEG solution by nasogastric tube, with one fatality [40, 41]. In
each case, the patient had emesis, suggesting aspiration of PEG. PEG also may
delay gastric emptying [42]. The strongest evidence for clinical benefit exists for
PEG, which has been evaluated in 19 trials, mostly compared to placebo, followed
by lactulose and tegaserod [31, 43]. In the largest study (304 patients treated for
6 months), patients given PEG had more bowel movements per week than patients
of placebo group [44]. In a retrospective series study, its efficacy was maintained for
up to 24 months [44, 45]. Patients prefer PEG preparations without electrolyte sup-
plements [46], which are necessary when a large volume is required [47].
electrolytes into the lumen and stimulate myenteric plexuses to induce intestinal
motility. The anthraquinones typically permit defecation 6–8 h after ingestion [51]
(Table 16.1). Senna has been shown to soften stools [52] and increase stool fre-
quency and weight. Anthraquinones cause apoptosis of colonic epithelial cells,
which are then phagocytosed by macrophages and appear as a lipofuscin-like pig-
ment that darkens the colonic mucosa, a condition termed Melanosis coli [53]
(Table 16.1). Moreover, these laxatives don’t induce significant adverse functional
or structural changes in the intestine. Animal studies have shown neither damage to
the myenteric plexus after long-term administration of sennosides [54] nor a func-
tional defect in motility [55].
stool form and had increased quality-of-life scores compared with those who
received placebo. Diarrhea was reported by 32% of patients who received the
sodium picosulfate [59] (Table 16.1).
A systematic review and meta-analysis of pharmacotherapies for chronic consti-
pation showed that bisacodyl and sodium picosulfate met the primary endpoints of
responder analysis with greater than or equal to 3 CSBM per week and an increase
over baseline of greater than or equal to 1 CSBM per week [60]. However, bisacodyl
may be superior to other prescription drugs in secondary endpoints, including an
increase from baseline in the number of spontaneous bowel movements (SBM) per
week and in the number of CSBM per week [17]. No studies have compared bisaco-
dyl to placebo in elderly patients with constipation [26].
Castor oil is obtained from the castor bean. After oral ingestion, it is hydrolyzed by
lipase in the small intestine to ricinoleic acid, which inhibits intestinal water absorp-
tion and stimulates intestinal motor function by damaging mucosal cells and releas-
ing neurotransmitters [61]. Cramping is frequent, and consequently castor oil is not
commonly used in clinical practice.
Several agents have been studied or are undergoing further studies for the treatment
of chronic constipation. These include colonic secretagogues, opioid antagonists,
and 5HT4 receptor agonists (Fig. 16.1 and Table 16.1).
Lubiprostone is an oral bicyclic fatty acid that activates the type 2 chloride channels
on the intestinal epithelial cells, thus secreting chloride and water into the intestinal
lumen [14]. In two phase III studies of 4 weeks’ duration, lubiprostone 24 μg twice
daily significantly enhanced bowel movement frequency and relieved other
constipation-related symptoms compared with placebo [62]. In a subgroup analysis,
lubiprostone also demonstrated efficacy in older patients. It is best reserved for
patients with severe constipation in whom other approaches have been unsuccessful
[8]. Nausea was the most common reported adverse event, occurring in up to 31.7%
of patients and leading to discontinuation in 5% [63] (Table 16.1). Lubiprostone is
also approved in the USA for the treatment of opioid-induced constipation as well
as for women with IBS with constipation at a dose of 8 μg twice daily [17].
Linaclotide and plecanatide are guanylate cyclase C receptor (GCC) agonists
that stimulate intestinal fluid secretion and transit [64]. Linaclotide is a minimally
absorbed 14-amino-acid peptide that activates the guanylate cyclase C receptor on
208 G. Fava
Opioids are a drug family largely used to manage chronic pain in clinical practice.
Opioid-induced constipation (OIC) results from the agonist actions of opioid medi-
cations at μ-opioid receptors, which are abundant throughout the gastrointestinal
tract. This adverse event is often underrecognized and undertreated in the elderly
[67]. In elderly patients with OIC and chronic pain who do not respond to lifestyle
interventions or laxatives, treatment of symptoms of constipation may be obtained
using peripherally acting μ-opioid receptor antagonists (PAMORAs), such as alvi-
mopan, methylnaltrexone, naloxegol, naldemedine, or alvimopan for paralytic ileus
[68]. These opioid receptor antagonists act peripherally and do not cross the blood-
brain barrier, thus decreasing the constipating effects of opioids without impairing
their analgesic effects [69]. The availability of the new pharmacologic agents meth-
ylnaltrexone, naloxegol, and lubiprostone may offer more specific and effective
treatment options for elderly patients with OIC and noncancer pain. Moreover,
healthcare providers should consider that methylnaltrexone [70] and naloxegol [71],
because of their peripheral mechanism of action, do not interfere with opioid-
induced, centrally mediated analgesia when administered to patients with noncan-
cer pain and OIC [67]. The complex multifactorial nature of constipation and its
potentially negative impact in elderly patients with OIC and chronic pain, coupled
with the availability of newer pharmacologic agents that target the underlying
mechanisms of constipation (i.e., PAMORAs), and the locally acting secretagogue
lubiprostone, provide an opportunity for healthcare providers to better manage their
elderly patients with OIC and chronic pain without change in pain medications [67].
constipation. Among them, cisapride, was widely used and clinically effective [72]
but was withdrawn from the commercial market in July 2000 for inducing poten-
tially lethal cardiac dysrhythmias. Similarly, tegaserod, another partial 5-HT4 ago-
nist, was also withdrawn in 2007 because of its potential cardiovascular effects.
Prucalopride, a selective high affinity 5HT4 receptor agonist, is a benzofuran
derivative that accelerates colonic transit in healthy humans and patients with
chronic constipation [73]. In a dose of 1 and 4 mg once daily, prucalopride has been
shown to be superior to placebo in 4- and 12-week trials and safe and well tolerated
in patients aged 65 years or older [74, 75]. In clinical trials, prucalopride 2 mg pro-
vided comparable efficacy to 4 mg, and it is therefore the widely used dosage in
clinical practice. The dose of prucalopride can be titrated up based on clinical
response [8]. The most frequent adverse effects were headaches, nausea, and diar-
rhea (Table 16.1). No cardiovascular side effects have been observed to date with
prucalopride, nor any electrocardiographic abnormalities been reported. In addition,
in a study of elderly constipated patients in nursing homes, no differences in vital
signs, electrocardiograph parameters, or Holter-monitoring results were found in
patients receiving prucalopride and placebo. Approximately 88% of the patients had
a history of cardiovascular disease [76].
Enemas and suppositories should be used only as needed for constipation in the
older adult, i.e., after several days of constipation, in order to prevent fecal impac-
tion. Both stimulate evacuation by distention or chemical action toward the rectal
walls, or soften hard stools, or both [17]. Enemas, in particular, need to be used with
caution since the extravasation of the solution into the submucosa can induce seri-
ous damage to the rectal wall [17].
Hypertonic sodium phosphate enemas cause distention and stimulation of the rec-
tum. Meisel et al. showed that in normal subjects, a single hypertonic phosphate
enema caused disruption of the surface epithelium in 17 of 21 biopsy specimens
[77]. The damage was evident by scanning electron microscopy, showing patchy
denudation of the surface epithelium, exposure of the lamina propria, and absence
of goblet cells. In every case, the mucosa was endoscopically pathological but
returned to normal within 1 week [77]. Phosphate enemas are widely used, although
studies documenting their efficacy are lacking. The use of sodium phosphate ene-
mas for the treatment of constipation in older adults is contraindicated. In a retro-
spective series, the use of sodium phosphate enemas in this setting of patients (mean
age, 80 years) was followed by complications including hypotension and volume
depletion, hyperphosphatemia, hypo- or hyperkalemia, metabolic acidosis, severe
hypocalcemia, renal failure, and electrocardiogram changes (prolonged QT inter-
val) [78]. In January 2014, the US Food and Drug Administration (FDA) issued a
210 G. Fava
Saline, tap water, or soapsuds enemas exert their action by distending the rectum
and softening feces. Stool evacuation typically occurs 2–5 min following adminis-
tration. If utilized with small volume, they usually don’t induce damage; however,
large volumes can be accompanied by serious effects such as water intoxication
hyperphosphatemia and other electrolyte disturbances in case of enema retention.
Soapsuds enemas can cause rectal mucosal damage and necrosis [17] (Table 16.1).
Mineral oils alter the stool by undergoing emulsification into the stool mass and
facilitating lubrication for stool passage. Long-term use can cause intestinal malab-
sorption of fat-soluble vitamins, anal seepage, and lipoid pneumonia in patients
predisposed to aspiration of liquids [17].
16.13 Probiotics
16.14 Conclusion
References
1. Talley NJ, O’Keefe EA, Zinsmeister AR, Melton LJ 3rd. Prevalence of gastrointestinal symp-
toms in the elderly: a population-based study. Gastroenterology. 1992;102:895.
2. Talley NJ, Fleming KC, Evans JM, et al. Constipation in an elderly community: a study of
prevalence and potential risk factors. Am J Gastroenterol. 1996;91:19.
3. Everhart JE, Go VL, Johannes RS, et al. A longitudinal survey of self-reported bowel habits in
the United States. Dig Dis Sci. 1989;34:1153.
212 G. Fava
4. Whitehead WE, Drinkwater D, Cheskin LJ, et al. Constipation in the elderly living at home.
Definition, prevalence, and relationship to lifestyle and health status. J Am Geriatr Soc.
1989;37:423.
5. Donald IP, Smith RG, Cruikshank JG, et al. A study of constipation in the elderly living at
home. Gerontology. 1985;31:112.
6. Harari D, Gurwitz JH, Avorn J, et al. Bowel habit in relation to age and gender. Findings
from the National Health Interview Survey and clinical implications. Arch Intern Med.
1996;156:315.
7. Choung RS, Locke GR 3rd, Schleck CD, et al. Cumulative incidence of chronic constipation:
a population-based study 1988-2003. Aliment Pharmacol Ther. 2007;26:1521.
8. Rao SSC. Constipation in the older adult. Up to date literature review current through: Jan
2022. This topic last updated: May 20, 2020.
9. Lindeman RD, Romero LJ, Liang HC, et al. Do elderly persons need to be encouraged to drink
more fluids? J Gerontol A Biol Sci Med Sci. 2000;55:M361.
10. Anti M, Pignataro G, Armuzzi A, et al. Water supplementation enhances the effect of high-
fiber diet on stool frequency and laxative consumption in adult patients with functional consti-
pation. Hepatogastroenterology. 1998;45:727–32.
11. Heaton EA. The call to stool and its relationship to constipation: a community study. Eur J
Gastroenterol Hepatol. 1994;6:145.
12. Harari D. Constipation and fecal incontinence in old age. Brocklehurst’s textbook of geriatric
medicine and gerontology, 8th edition. 2017;105, 877–894.e5.
13. Rao SS, Kavelock R, Beaty J, et al. Effects of fat and carbohydrate meals on colonic motor
response. Gut. 2000;46:205.
14. Rao SS, Go JT. Update on the management of constipation in the elderly: new treatment
options. Clin Interv Aging. 2010;5:163.
15. Johnson CK, Kolasa K, Chenoweth W, et al. Health, laxation and food habit influences on fiber
intake of older women. J Am Diet Assoc. 1980;77:551–7.
16. Gallagher P, O’Mahony D. Constipation in old age. Best Pract Res Clin Gastroenterol.
2009;23:875.
17. Iturrino JC, Lembo AJ. Chapter 19. Constipation. In: Sleisenger and Fordtran’s gastrointestinal
and liver disease. 2020. p. 250–275.e8.
18. American College of Gastroenterology Chronic Constipation Task Force. An evidence- based
approach to the management of chronic constipation in North America. Am J Gastroenterol.
2005;100(Suppl 1):S1.
19. Ramkumar D, Rao SS. Efficacy and safety of traditional medical therapies for chronic consti-
pation: systematic review. Am J Gastroenterol. 2005;100:936.
20. Locke GR 3rd, Pemberton JH, Phillips SF. American Gastroenterological Association medical
position statement: guidelines on constipation. Gastroenterology. 2000;119:1761.
21. Donowitz M, Rood RP. Magnesium hydroxide: new insights into the mechanism of its laxative
effect and the potential involvement of prostaglandin E2. J Clin Gastroenterol. 1992;14:20–6.
22. Saris NE, Mervaala E, Karppanen H, et al. Magnesium. An update on physiological, clinical
and analytical aspects. Clin Chim Acta. 2000;294:1–26.
23. Izdes S, Kesimci E, Kanbak O. Paralytic ileus as a complication of iatrogenic hypermagnesae-
mia without renal dysfunction. Anaesth Intensive Care. 2008;36:124.
24. Phosphate, potassium and sodium. Source Lexycomp®.
25. Wright RA. Lactulose-induced megacolon. Gastrointest Endosc. 1988;34:489–90.
26. Kang SJ, Cho YS, Lee TH, Kim SE, Ryu HS, Kim JW, Park SY, Lee YJ, Shin JE, Constipation
Research Group of the Korean Society of Neurogastroenterology and Motility. Medical man-
agement of constipation in the elderly patients: systematic review. J Neurogastroenterol Motil.
2021;27(4):495–512.
27. Ruby CM, Fillenbaum GG, Kuchibhatla MN, Hanlon JT. Laxative use in the community-
dwelling elderly. Am J Geriatr Pharmacother. 2003;1:11.
28. Harari D, Gurwitz JH, Avorn J, et al. Constipation: assessment and management in an institu-
tionalized elderly population. J Am Geriatr Soc. 1994;42:947.
16 Constipation 213
29. Petticrew M, Watt I, Sheldon T. Systematic review of the effectiveness of laxatives in the
elderly. Health Technol Assess. 1997;1:i–iv, 1–52.
30. Fleming V, Wade WE. A review of laxative therapies for treatment of chronic constipation in
older adults. Am J Geriatr Pharmacother. 2010;8:514–50.
31. Ford A, Suares N. Effect of laxatives and pharmacological therapies in chronic idiopathic con-
stipation: systematic review and meta-analysis. Gut. 2011;60:209–18. [PubMed: 21205879].
32. Wesselius-De Casparis A, Braadbaart S, Bergh-Bohlken GE, Mimica M. Treatment of chronic
constipation with lactulose syrup: results of a double-blind study. Gut. 1968;9:84–6.
33. Sanders JF. Lactulose syrup assessed in a double-blind study of elderly constipated patients. J
Am Geriatr Soc. 1978;26:236–9.
34. Vanderdonckt J, Coulon J, Denys W, Ravelli GP. Study of the laxative effect of lactitol (impor-
talr) in an elderly institutionalized, but not bedridden, population suffering from chronic con-
stipation. J Clin Exp Gerontol. 1990;12:171–89.
35. Hyams JS. Sorbitol intolerance: an unappreciated cause of functional gastrointestinal com-
plaints. Gastroenterology. 1983;84:30–3.
36. Lederle FA, Busch DL, Mattox KM, et al. Cost-effective treatment of constipation in
the elderly: a randomized double-blind comparison of sorbitol and lactulose. Am J Med.
1990;89:597–601.
37. Corazziari E, Badiali D, Habib FI, et al. Small volume isosmotic polyethylene glycol electro-
lyte balanced solution (PMF-100) in treatment of chronic nonorganic constipation. Dig Dis
Sci. 1996;41:1636–42.
38. Garlehner G, Jonas DE, Morgan LC, et al. Drug class review on constipation drugs. Portland:
Oregon Health & Science University; 2007.
39. Belsey J, Epstein O, Heresbach D. Systematic review: adverse event reports for oral sodium
phosphate and polyethylene glycol. Aliment Pharmacol Ther. 2009;29:15–28.
40. Argent A, Hatherill M, Reynolds L, Purves L. Fulminant pulmonary oedema after administra-
tion of a balanced electrolyte polyethylene glycol solution. Arch Dis Child. 2002;86:209.
41. Marschall HU, Bartels F. Life-threatening complications of nasogastric administration of poly-
ethylene glycol-electrolyte solutions (Go-lytely) for bowel cleansing. Gastrointest Endosc.
1998;47:408–10.
42. Coremans G, Vos R, Margaritis V, et al. Small doses of the unabsorbable substance polyeth-
ylene glycol 3350 accelerate oro-caecal transit, but slow gastric emptying in healthy subjects.
Dig Liver Dis. 2005;37:97–101.
43. Pare P, Fedorak RN. Systematic review of stimulant and nonstimulant laxatives for the treat-
ment of functional constipation. Can J Gastroenterol Hepatol. 2014;28:549–57.
44. DiPalma JA, Cleveland MV, McGowan J, et al. A randomized, multicenter, placebo-controlled
trial of polyethylene glycol laxative for chronic treatment of chronic constipation. Am J
Gastroenterol. 2007;102:1436–41.
45. Migeon-Duballet I, Chabin M, Gautier A, et al. Long-term efficacy and cost-effectiveness of
polyethylene glycol 3350 plus electrolytes in chronic constipation: a retrospective study in a
disabled population. Curr Med Res Opin. 2006;22:1227–35.
46. Szojda MM, Mulder CJJ, Felt-Bersma RJF. Differences in taste between two polyethylene
glycol preparations. J Gastrointest Liver Dis. 2007;16:379–81.
47. DiPalma JA, Smith JR, Cleveland MV. Overnight efficacy of polyethylene glycol laxative. Am
J Gastroenterol. 2002;97:1776–9.
48. Bharucha AE, Lacy BE. Mechanisms, evaluation, and management of chronic constipation.
Gastroenterology. 2020;158(5):1232–1249.e3.
49. Ikarashi N, Baba K, Ushiki T, et al. The laxative effect of bisacodyl is attributable to decreased
aquaporin-3 expression in the colon induced by increased PGE2 secretion from macrophages.
Am J Physiol Gastrointest Liver Physiol. 2011;301:G887–95.
50. Manabe N, Cremonini F, Camilleri M, et al. Effects of bisacodyl on ascending colon emptying
and overall colonic transit in healthy volunteers. Aliment Pharmacol Ther. 2009;30:930–6.
51. Neims DM, McNeill J, Giles TR, Todd F. Incidence of laxative abuse in community and
bulimic populations: a descriptive review. Int J Eat Disord. 1995;17:211–28.
214 G. Fava
52. Exton-Smith AN, Bendall MJ, Kent F. A new technique for measuring the consistency of
faeces: a report on its application to the assessment of Senokotot therapy in the elderly. Age
Ageing. 1975;4:58–62.
53. Badiali D, Marcheggiano A, Pallone F, et al. Melanosis of the rectum in patients with chronic
constipation. Dis Colon Rectum. 1985;28:241–5.
54. Rudolph RL, Mengs U. Electron microscopical studies on rat intestine after long-term treat-
ment with sennosides. Pharmacology. 1988;36(Suppl. 1):188–93.
55. Fioramonti J, Dupuy C, Buéno L. In vivo motility of rat colon chronically pretreated with
sennosides. Pharmacology. 1993;47(Suppl. 1):155–61.
56. Kamm MA, Lennard-Jones JE, Thompson DG, et al. Dynamic scanning defines a colonic
defect in severe idiopathic constipation. Gut. 1988;29:1085–92.
57. Mengs U, Rudolph RL. Light and electron-microscopic changes in the colon of the Guinea pig
after treatment with anthranoid and non-anthranoid laxatives. Pharmacology. 1993;47(Suppl.
1):172–7.
58. Kamm MA, Mueller-Lissner S, Wald A, Richter E, Swallow R, Gessner U. Oral bisacodyl is
effective and well-tolerated in patients with chronic constipation. Clin Gastroenterol Hepatol.
2011;9(7):577. Epub 2011 Mar 25.
59. Mueller-Lissner S, Kamm MA, Wald A, et al. Multicenter, 4-week, double-blind, random-
ized, placebo-controlled trial of sodium picosulfate in patients with chronic constipation. Am
J Gastroenterol. 2010;105:897–903.
60. Nelson AD, Camilleri M, Chirapongsathorn S, et al. Comparison of efficacy of pharmaco-
logical treatments for chronic idiopathic constipation: a systematic review and network meta-
analysis. Gut. 2017;66:1611–22.
61. van Gorkom BA, de Vries EG, Karrenbeld A, Kleibeuker JH. Review article: anthranoid laxa-
tives and their potential carcinogenic effects. Aliment Pharmacol Ther. 1999;13:443–52.
62. Johanson JF, Morton D, Geenen J, Ueno R. Multicenter, 4-week, double-blind, randomized,
placebo-controlled trial of lubiprostone, a locally-acting type-2 chloride channel activator, in
patients with chronic constipation. Am J Gastroenterol. 2008;103(1):170. Epub 2007 Oct 4.
63. Rohack JJ, Mehta BR, Subramanyam K. Hyperphosphatemia and hypocalcemic coma associ-
ated with phosphate enema. South Med J. 1985;78:1241–2.
64. Lembo AJ, Kurtz CB, Macdougall JE, Lavins BJ, Currie MG, Fitch DA, Jeglinski BI,
Johnston JM. Efficacy of linaclotide for patients with chronic constipation. Gastroenterology.
2010;138(3):886. Epub 2010 Jan 4.
65. Eutamene H, Bradesi S, Larauche M, et al. Guanylate cyclase C-mediated antinocicep-
tive effects of linaclotide in rodent models of visceral pain. Neurogastroenterol Motil.
2010;22:312–e84.
66. Schoenfeld P, Lacy BE, Chey WD, Lembo AJ, Kurtz CB, Reasner DS, Bochenek W, Tripp
K, Currie MG, Fox SM, Blakesley RE, OʼDea CR, Omniewski ND, Hall ML. Low-dose
Linaclotide (72 μg) for chronic idiopathic constipation: a 12-week, randomized, double-blind,
placebo-controlled trial. Am J Gastroenterol. 2018;113(1):105. Epub 2017 Aug 22.
67. Chokhavatia S, John ES, Bridgeman MB, Dixit D. Constipation in elderly patients with non-
cancer pain: focus on opioid-induced constipation. Drugs Aging. 2016;33:557–74. https://doi.
org/10.1007/s40266-016-0381-2.
68. Crockett SD, Greer KB, Heidelbaugh JJ, Falck-Ytter Y, Hanson BJ, Sultan S, American
Gastroenterological Association Institute Clinical Guidelines Committee. American
Gastroenterological Association Institute guideline on the medical management of opioid-
induced constipation. Gastroenterology. 2019;156(1):218. Epub 2018 Oct 16.
69. Nelson AD, Camilleri M. Chronic opioid induced constipation in patients with nonmalignant
pain: challenges and opportunities. Ther Adv Gastroenterol. 2015;8:206–20.
70. Thomas J, Karver S, Cooney GA, et al. Methylnaltrexone for opioid-induced constipation in
advanced illness. N Engl J Med. 2008;358:2332–43.
71. Chey WD, Webster L, Sostek M, et al. Naloxegol for opioid-induced constipation in patients
with noncancer pain. N Engl J Med. 2014;370:2387–96.
16 Constipation 215
72. Müller-Lissner SA. Treatment of chronic constipation with cisapride and placebo. Gut.
1987;28:1033–8.
73. Bouras EP, Camilleri M, Burton DD, et al. Prucalopride accelerates gastrointestinal and colonic
transit in patients with constipation without a rectal evacuation disorder. Gastroenterology.
2001;120:354–60.
74. Quigley EM, Vandeplassche L, Kerstens R, Ausma J. Clinical trial: the efficacy, impact on
quality of life, and safety and tolerability of prucalopride in severe chronic constipation—
a 12-week, randomized, double-blind, placebo-controlled study. Aliment Pharmacol Ther.
2009;29(3):315. Epub 2008 Nov 8.
75. Müller-Lissner S, Rykx A, Kerstens R, Vandeplassche L. A double-blind, placebo-controlled
study of prucalopride in elderly patients with chronic constipation. Neurogastroenterol Motil.
2010;22(9):991.
76. Camilleri M, Beyens G, Kerstens R, et al. Safety assessment of prucalopride in elderly patients
with constipation: a double-blind, placebo-controlled study. Neurogastroenterol Motil.
2009;21:1256–e117.
77. Meisel JL, Bergman D, Graney D, et al. Human rectal mucosa: proctoscopic and morphologi-
cal changes caused by laxatives. Gastroenterology. 1977;72:1274–9.
78. Ori Y, Rozen-Zvi B, Chagnac A, Herman M, Zingerman B, Atar E, Gafter U, Korzets
A. Fatalities and severe metabolic disorders associated with the use of sodium phosphate ene-
mas: a single center’s experience. Arch Intern Med. 2012;172(3):263.
79. FDA warns of possible harm from exceeding recommended dose of over-the-counter sodium
phosphate products to treat constipation. http://www.fda.gov/Drugs/DrugSafety/ucm380757.
htm. Accessed 8 Jan 2014.
80. Mendoza J, Legido J, Rubio S, Gisbert JP. Systematic review: the adverse effects of sodium
phosphate enema. Aliment Pharmacol Ther. 2007;26(1):9.
81. Flig E, Hermann TW, Zabel M. Is bisacodyl absorbed at all from suppositories in man? Int J
Pharm. 2000;196:11–20.
82. Larkin PJ, Cherny NI, La Carpia D, et al. Diagnosis, assessment and management of consti-
pation in advanced cancer: ESMO clinical practice guidelines. Ann Oncol. 2018;29 Suppl
4(Suppl 4):iv111–25.
83. Gandell D, Straus SE, Bundookwala M, Tsui V, Alibhai SMH. Treatment of constipation in
older people. CMAJ. 2013;185(8):663–70.
84. McRorie JW, Daggy BP, Morel JG, et al. Psyllium is superior to docusate sodium for treatment
of chronic constipation. Aliment Pharmacol Ther. 1998;12:491–7.
85. von Maltzahn M, Tanzini RM, Leu R, Wong CL. When less may be more: discontinuing docu-
sate from a standardized hospital order set. J Geriatr Oncol. 2022;13:111–3.
86. Martínez-Martínez MI, Calabuig-Tolsá R, Cauli O. The effect of probiotics as a treatment for
constipation in elderly people: a systematic review. Arch Gerontol Geriatr. 2017;71:142–9.
87. Aoyagi Y, et al. Independent and interactive effects of habitually ingesting fermented milk
products containing Lactobacillus casei strain Shirota and of engaging in moderate habitual
daily physical activity on the intestinal health of older people. Front Microbiol. 2019;10:1477.
88. Matsumoto K. Modification of gastrointestinal function: improvement of diarrhea and consti-
pation. In: Lactobacillus casei strain Shirota—intestinal microbiota and human health: new
advances—Supplementary Edition, ed. Yakult Central Institute for Microbiological Research
Tokyo: Yakult Honsha Co., Ltd. 2013. p. 41–51.
89. Mai TT, Hop DV, Anh TTV, Lam NT. Recovery of Lactobacillus casei strain Shirota (LcS)
from the intestine of healthy Vietnamese adults after intake of fermented milk. Asia Pac J Clin
Nutr. 2017;26:72–7.
Pain
17
Sophie Pautex, Monica Escher, and Petra Vayne-Bossert
17.1 Introduction
The International Association for the Study of Pain (IASP) definition states that
pain is an unpleasant sensory and emotional experience associated with actual or
potential tissue damage or described in terms of such damage. Although aging and
especially neurocognitive disorders are associated with structural, biochemical, and
functional changes in the nervous system, there are no arguments that these changes
have a clear and important influence on the perception of pain in the older patient
population [1]. Chronic pain, i.e. pain present for more than 3–6 months, affects
25–76% of home-dwelling people over 65 years and up to 93% of older people in
nursing homes [2, 3]. Most common causes are musculoskeletal pain (up to 83% of
patients) such as joint pain and low back pain due to osteoarthritis, pain from previ-
ous fractures, and peripheral neuropathies. Older adults have often multiple pain-
associated conditions that likely reflect multiple physiological mechanisms for pain.
Linking pain qualities with other associated pain characteristics serve to develop a
multidimensional approach to geriatric pain assessment [4]. In most studies, about
60% of pain is of pure nociceptive origin resulting from an actual or threatened
damage to nonneural tissue and is due to the activation of nociceptors located in
somatic (e.g., skin, musculoskeletal tissues) or visceral tissues. Secondly, pain can
be caused by a lesion or disease of the nervous system which is called neuropathic
pain. Pure neuropathic pain is present in about 10% of the older population, while
combined nociceptive and neuropathic pain is found in about 1 out of 3 older per-
sons. Persistent pain has a significant impact on all levels of functioning and on
quality of life. Inadequate treatment is associated with various adverse outcomes in
older people, including functional impairment, falls, slow rehabilitation, mood
changes (depression and anxiety), decreased socialization, sleep and appetite distur-
bance, and greater healthcare use and costs [5, 6]. The management of this persis-
tent pain should be more multidimensional than for acute pain, in particular in this
population.
Pain management includes nonpharmacologic and pharmacologic approaches,
and shared decision making is essential to balance their benefits and burdens. Pain
management goals should be determined with patients before the initiation of any
treatment, and there must be ongoing monitoring of treatment efficacy and adverse
effects over time. Interventions should generally be targeted at improvements in
pain-related disability rather than pain intensity, because improvements in disability
are more tangible outcomes among persons with persistent pain [7, 8].
17.2.1 Paracetamol
NSAID-related side effects as the cause for hospitalization in 23.5% of patients [14,
15]. Gastrointestinal toxicity due to NSAIDs is dose dependent, increases with age
and the concomitant use of cardioprotective doses of aspirin, which is a common
medication in this group. Use of NSAIDs can also lead to renal impairment through
sodium and water retention, electrolyte imbalances, prerenal azotemia, and reduced
blood flow to the kidneys. The concomitant use of NSAIDS and several medications
commonly prescribed in older adults (i.e., anticoagulants, aspirin, and selective
serotonin reuptake inhibitors) increases the risk of bleeding.
Topical NSAIDs are often used for knee or hand osteoarthritis (OA)-related pain.
Evidence from a recent randomized controlled trial showed the comparable efficacy
of topical diclofenac sodium and oral NSAIDs in the treatment of knee osteoarthri-
tis with fewer adverse effects [16]. The mechanism is thought to be an increased
delivery of the drug to the adjacent synovium without significant systemic
absorption.
17.2.3 Opioids
Opioid analgesics are considered for managing severe pain or failed response to
other treatments in older patients. Although the short-term efficacy of opioid anal-
gesics in chronic pain in this group is established, limited evidence is available
regarding the long-term efficacy because of the discontinuation of opioid therapy
due to various poorly tolerated side effects. A prerequisite before initiation of opioid
therapy is the evaluation of risk versus benefit. While taking into account the adverse
outcomes, mandatory efforts should be made to reduce the risks. Administration of
opioids in the older patient is done on a trial basis to titrate the effective dose reach-
ing the therapeutic goal with minimal adverse effects, starting with the lowest pos-
sible dose and continuing with gradual titration. Patients with moderate to severe
pain and diminished pain-related quality of life should be considered for opioid
therapy (low quality of evidence, strong recommendation) [17]. Studies comparing
the efficacy and tolerability of opioids, such as transdermal fentanyl, oral morphine,
and sublingual or transdermal buprenorphine in the older population and other pop-
ulations have shown that older people respond to opioid treatment as well as, or
even better than, younger age groups [18–20]. The “start low and go slow” approach
is essential when dosing opioids. Patients who report severe pain or those who have
experienced uncontrolled pain for prolonged periods of time will likely require
ongoing titration of opioid therapy to balance pain relief with adverse effects.
Opioids should be started at 25–50% of the recommended dose for younger
adults [21].
The decreased volume of distribution that occurs owing to decreased total body
water with aging may also result in increased plasma levels of more hydrophilic
opioids (e.g., morphine) compared with levels observed in younger persons. In gen-
eral, oral bioavailability does not seem to be affected by age, and although first-pass
metabolism may be affected, dosage adjustments are not routinely necessary beyond
the 25–50% dose reduction recommended at opioid initiation [22, 23]. Decrements
220 S. Pautex et al.
Misuse or abuse: In recent years, the abuse or misuse of opioids has steadily
gone up, especially in the USA [33]. The older population is also concerned although
much less than the younger adults. Nevertheless, clinicians must remain vigilant
about the possibility of misuse or abuse of opioid agents in older adults and apply
the abovementioned precautions while prescribing and initiating these agents in
patients with non-cancer pain [34].
Respiratory depression is more frequent (although still rare) in patients who are
opioid naive, receive the agent intravenously, and have increased their doses rapidly.
Furthermore, patients receiving drugs with variable pharmacokinetics such as meth-
adone, patients with concomitant use of central nervous system depressing drugs
such as benzodiazepines or barbiturates, or patients who develop sudden acute kid-
ney failure are more at risk for this adverse event.
Tramadol
Tramadol, a weak mu-opioid agonist with additional serotonin and norepinephrine
reuptake inhibition, is not routinely recommended for older adults with moderate to
severe pain but is commonly used. Importantly, its mu-opioid receptor activity
results in a similar side effect profile as other opioids and requires similar cautions
described herein. Nausea, dizziness, and tiredness seem particularly common [35].
In addition, tramadol increases seizure risk, particularly at doses higher than
300 mg/day, the maximum daily dose recommended in an older adult. Lastly, the
development of serotonin syndrome may occur with the use of tramadol, particu-
larly with concomitant use of serotonergic drugs. Tramadol is activated to its opioid
metabolite by CYP2D6. Patients should be closely monitored to detect a potential
clinical impact—either decreased efficacy or increased side effects—due to drug-
drug interactions or to modified metabolism in ultrarapid and poor metabolizers.
Tramadol should be initiated at 25 mg three or four times daily and increased in
25 mg increments every 2–3 days to an initial goal of 100 mg–200 mg/day.
Tapentadol
Tapentadol is a weak opioid that acts on the mu receptor and on neuronal reuptake
of noradrenaline. Unlike tramadol, it has only weak effects on serotonin reuptake. It
has no active metabolites and does not need to be activated to exert its analgesic
opioid effect [36]. There are no sufficient data to support the use of newer drug
tapentadol over other opioids.
Morphine
Morphine has been used to treat malignant pain for many years and has been the
subject of a large number of trials, generally involving small numbers of patients.
Morphine is metabolized in the liver by glucuronidation. Morphine-6-glucuronide
(M6G) is a potent analgesic, and morphine-3-glucuronide (M3G) may cause neuro-
excitatory effects [37]. Renal excretion accounts for about 90% of the elimination
222 S. Pautex et al.
Fentanyl
Fentanyl, a phenylpiperidine derivative, is chemically related to pethidine. It is a
pure fat-soluble μ-opioid agonist. Its volume of distribution increases in older
patients because of the increased fat-to-lean body mass ratio that accompanies
224 S. Pautex et al.
aging, hence prolonging its effective half-life. Fentanyl can be used in renal failure.
It makes it an interesting alternative to morphine as renal function is altered with
age and older patients are more likely to experiment acute renal failure in case of
dehydration or nephrotoxic drugs use.
Buprenorphine
Buprenorphine is a semisynthetic, partial μ-opioid agonist. It also weakly binds as
an antagonist at kappa and delta receptors. It has high affinity for μ-receptors and is
not readily reversed by naloxone. Buprenorphine is excreted primarily in the feces
unchanged and is considered safe in persons with renal impairment [39]. Because of
significant first-pass effect, it is available in sublingual form or transdermal patch.
The sublingual formulation is of limited utility in patients with cognitive impair-
ment or dry mouth since the tablet must be kept under the tongue until it is dissolved
completely.
Methadone
Methadone is a synthetic lipophilic opioid analgesic. It is extensively metabolized
by CYP450 enzymes (CYP3A4, CYP2B6, CYP2C19, CYP2C9, and CYP2D6) into
inactive metabolites, and it is considered safe for use in persons with renal impair-
ment. However, equianalgesic ratios between morphine and methadone are dose
dependent, the half-life is highly variable, and numerous drug interactions must be
considered. Therefore, methadone should be used only by practitioners experienced
with this agent [40].
17.2.5 Cannabinoids
The efficacy and safety profile of cannabinoids are not established in this popula-
tion. Common side effects include euphoria, anxiety, psychosis, sedation, dizziness,
cognitive effects, tachycardia, palpitations, and postural hypotension. Due to a nar-
row therapeutic index, older patients are at higher risk of dysphoric effects of can-
nabinoids [47].
In conclusion, as the quality of research is weak, and few older patients have
been enrolled in cannabinoid studies, dedicated research is needed to determine the
efficiency and safety of cannabinoids in older patients.
References
1. Gibson SJ, Farrell M. A review of age differences in the neurophysiology of nociception and
the perceptual experience of pain. Clin J Pain. 2004;20(4):227–39.
2. American Geriatrics Society Panel on Pharmacological Management of Persistent Pain in
Older Persons. Pharmacological management of persistent pain in older persons. J Am Geriatr
Soc. 2009;57(8):1331–46.
3. AGS Panel on Persistent Pain in Older Persons. The management of persistent pain in older
persons. J Am Geriatr Soc. 2002;50(6 Suppl):S205–24.
4. Thakral M, Shi L, Foust JB, Patel KV, Shmerling RH, Bean JF, et al. Pain quality descriptors
in community-dwelling older adults with nonmalignant pain. Pain. 2016;157(12):2834–42.
5. Sawyer P, Bodner EV, Ritchie CS, Allman RM. Pain and pain medication use in community-
dwelling older adults. Am J Geriatr Pharmacother. 2006;4(4):316–24.
6. Won AB, Lapane KL, Vallow S, Schein J, Morris JN, Lipsitz LA. Persistent nonmalignant
pain and analgesic prescribing patterns in elderly nursing home residents. J Am Geriatr Soc.
2004;52(6):867–74.
7. Bernabei R, Gambassi G, Lapane K, Landi F, Gatsonis C, Dunlop R, et al. Management of
pain in elderly patients with cancer. SAGE Study Group. Systematic assessment of geriatric
drug use via epidemiology. JAMA. 1998;279(23):1877–82.
226 S. Pautex et al.
8. Landi F, Onder G, Cesari M, Gambassi G, Steel K, Russo A, et al. Pain management in frail,
community-living elderly patients. Arch Intern Med. 2001;161(22):2721–4.
9. Lehti TE, Rinkinen MO, Aalto U, Roitto HM, Knuutila M, Ohman H, et al. Prevalence of mus-
culoskeletal pain and analgesic treatment among community-dwelling older adults: changes
from 1999 to 2019. Drugs Aging. 2021;38(10):931–7.
10. Mian P, Allegaert K, Spriet I, Tibboel D, Petrovic M. Paracetamol in older people: towards
evidence-based dosing? Drugs Aging. 2018;35(7):603–24.
11. Pickering G, Schneider E, Papet I, Pujos-Guillot E, Pereira B, Simen E, et al. N metabo-
lism after major surgery: a greater challenge with increasing age. Clin Pharmacol Ther.
2011;90(5):707–11.
12. Graham GG, Davies MJ, Day RO, Mohamudally A, Scott KF. The modern pharmacology of
paracetamol: therapeutic actions, mechanism of action, metabolism, toxicity and recent phar-
macological findings. Inflammopharmacology. 2013;21(3):201–32.
13. da Costa BR, Reichenbach S, Keller N, Nartey L, Wandel S, Jüni P, et al. Effectiveness of
non-steroidal anti-inflammatory drugs for the treatment of pain in knee and hip osteoarthritis:
a network meta-analysis. Lancet. 2017;390(10090):e21–33.
14. Trelle S, Reichenbach S, Wandel S, Hildebrand P, Tschannen B, Villiger PM, et al.
Cardiovascular safety of non-steroidal anti-inflammatory drugs: network meta-analysis.
BMJ. 2011;342:c7086.
15. Wegman A, van der Windt D, van Tulder M, Stalman W, de Vries T. Nonsteroidal antiinflam-
matory drugs or acetaminophen for osteoarthritis of the hip or knee? A systematic review of
evidence and guidelines. J Rheumatol. 2004;31(2):344–54.
16. Tugwell PS, Wells GA, Shainhouse JZ. Equivalence study of a topical diclofenac solution
(pennsaid) compared with oral diclofenac in symptomatic treatment of osteoarthritis of the
knee: a randomized controlled trial. J Rheumatol. 2004;31(10):2002–12.
17. Kaye AD, Baluch A, Scott JT. Pain management in the elderly population: a review. Ochsner
J. 2010;10(3):179–87.
18. Kaiko RF. Age and morphine analgesia in cancer patients with postoperative pain. Clin
Pharmacol Ther. 1980;28(6):823–6.
19. Menten J, Desmedt M, Lossignol D, Mullie A. Longitudinal follow-up of TTS-fentanyl use in
patients with cancer-related pain: results of a compassionate-use study with special focus on
elderly patients. Curr Med Res Opin. 2002;18(8):488–98.
20. Nasar MA, McLeavy MA, Knox J. An open study of sub-lingual buprenorphine in the treat-
ment of chronic pain in the elderly. Curr Med Res Opin. 1986;10(4):251–5.
21. Mercadante S, Giarratano A. Assessing age and gender in studies of breakthrough pain medi-
cations. Curr Med Res Opin. 2014;30(7):1353–6.
22. Tegeder I, Geisslinger G, Lotsch J. [Therapy with opioids in liver or renal failure]. Schmerz.
1999;13(3):183–195.
23. Tegeder I, Lotsch J, Geisslinger G. Pharmacokinetics of opioids in liver disease. Clin
Pharmacokinet. 1999;37(1):17–40.
24. Dean M. Opioids in renal failure and dialysis patients. J Pain Symptom Manag.
2004;28(5):497–504.
25. Murtagh FE, Chai MO, Donohoe P, Edmonds PM, Higginson IJ. The use of opioid analgesia
in end-stage renal disease patients managed without dialysis: recommendations for practice. J
Pain Palliat Care Pharmacother. 2007;21(2):5–16.
26. Cherny N, Ripamonti C, Pereira J, Davis C, Fallon M, McQuay H, et al. Strategies to manage the
adverse effects of oral morphine: an evidence-based report. J Clin Oncol. 2001;19(9):2542–54.
27. Miller M, Sturmer T, Azrael D, Levin R, Solomon DH. Opioid analgesics and the risk of frac-
tures in older adults with arthritis. J Am Geriatr Soc. 2011;59(3):430–8.
28. Rolita L, Spegman A, Tang X, Cronstein BN. Greater number of narcotic analgesic prescrip-
tions for osteoarthritis is associated with falls and fractures in elderly adults. J Am Geriatr Soc.
2013;61(3):335–40.
29. Solomon DH, Rassen JA, Glynn RJ, Garneau K, Levin R, Lee J, et al. The comparative safety
of opioids for nonmalignant pain in older adults. Arch Intern Med. 2010;170(22):1979–86.
17 Pain 227
30. Morrison RS, Magaziner J, Gilbert M, Koval KJ, McLaughlin MA, Orosz G, et al. Relationship
between pain and opioid analgesics on the development of delirium following hip fracture. J
Gerontol A Biol Sci Med Sci. 2003;58(1):76–81.
31. Caraceni A, Hanks G, Kaasa S, Bennett MI, Brunelli C, Cherny N, et al. Use of opioid analge-
sics in the treatment of cancer pain: evidence-based recommendations from the EAPC. Lancet
Oncol. 2012;13(2):e58–68.
32. Hanks GW, Conno F, Cherny N, Hanna M, Kalso E, McQuay HJ, et al. Morphine and alterna-
tive opioids in cancer pain: the EAPC recommendations. Br J Cancer. 2001;84(5):587–93.
33. Shoff C, Yang TC, Shaw BA. Trends in opioid use disorder among older adults: analyzing
Medicare data, 2013-2018. Am J Prev Med. 2021;60(6):850–5.
34. Edlund MJ, Steffick D, Hudson T, Harris KM, Sullivan M. Risk factors for clinically recog-
nized opioid abuse and dependence among veterans using opioids for chronic non-cancer pain.
Pain. 2007;129(3):355–62.
35. Toupin April K, Bisaillon J, Welch V, Maxwell LJ, Juni P, Rutjes AW, et al. Tramadol for osteo-
arthritis. Cochrane Database Syst Rev. 2019;5:CD005522.
36. Veal FC, Peterson GM. Pain in the frail or elderly patient: does Tapentadol have a role? Drugs
Aging. 2015;32(6):419–26.
37. Lee J, Lakha SF, Mailis A. Efficacy of low-dose oral liquid morphine for elderly patients
with chronic non-cancer pain: retrospective chart review. Drugs Real World Outcomes.
2015;2(4):369–76.
38. Gianni W, Ceci M, Bustacchini S, Corsonello A, Abbatecola AM, Brancati AM, et al. Opioids
for the treatment of chronic non-cancer pain in older people. Drugs Aging. 2009;26(Suppl
1):63–73.
39. Vadivelu N, Hines RL. Management of chronic pain in the elderly: focus on transdermal
buprenorphine. Clin Interv Aging. 2008;3(3):421–30.
40. Lugo RA, Satterfield KL, Kern SE. Pharmacokinetics of methadone. J Pain Palliat Care
Pharmacother. 2005;19(4):13–24.
41. Pickering G, Marcoux M, Chapiro S, David L, Rat P, Michel M, et al. An algorithm for neuro-
pathic pain management in older people. Drugs Aging. 2016;33(8):575–83.
42. Haslam C, Nurmikko T. Pharmacological treatment of neuropathic pain in older persons. Clin
Interv Aging. 2008;3(1):111–20.
43. Wiffen PJ, Derry S, Moore RA, Aldington D, Cole P, Rice AS, et al. Antiepileptic drugs for
neuropathic pain and fibromyalgia—an overview of Cochrane reviews. Cochrane Database
Syst Rev. 2013;(11):CD010567.
44. Goldstein DJ, Lu Y, Detke MJ, Lee TC, Iyengar S. Duloxetine vs. placebo in patients with
painful diabetic neuropathy. Pain. 2005;116(1–2):109–18.
45. Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, et al.
Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis.
Lancet Neurol. 2015;14(2):162–73.
46. Moore RA, Derry S, Aldington D, Cole P, Wiffen PJ. Amitriptyline for neuropathic pain in
adults. Cochrane Database Syst Rev. 2015;(7):CD008242.
47. Pisani S, McGoohan K, Velayudhan L, Bhattacharyya S. Safety and tolerability of natural and
synthetic cannabinoids in older adults: a systematic review and meta-analysis of open-label
trials and observational studies. Drugs Aging. 2021;38:887.
Hypertension
18
Timo E. Strandberg, Mirko Petrovic, and Athanase Benetos
Hypertension is a leading cause of disability and major risk factor for cardiovascu-
lar diseases (CVD), which are common among older people, such as stroke, heart
failure, and coronary heart disease. Hypertension is also an important risk factor for
cognitive decline. In modern societies, systolic blood pressure generally starts to
rise after 50 years of age, and consequently systolic hypertension and antihyperten-
sive treatment are very prevalent among older people.
The most recent meta-analysis of randomized controlled trials (RCT) of pharma-
cotherapy clearly demonstrates that antihypertensive treatment is effective in the
prevention of CVD irrespective of older age and blood pressure level at randomiza-
tion [1]. However, evidence from age 85 and older is weaker, probably because of a
relatively small number of oldest-old participants in trials. Besides prevention of
CVD, it is noteworthy that according to systematic reviews of RCTs, medical treat-
ment of hypertension is actually the only method proven to prevent adjudicated
incident dementia—not only cognitive decline [2–4].
Results from RCTs also show that besides chronological age, strict blood pres-
sure goals are irrelevant [1] and the benefit from treatment depends on the overall
risk of the patient. Because “physiologically” normal blood pressure is as low as
T. E. Strandberg (*)
University of Helsinki and Helsinki University Hospital, Helsinki, Finland
University of Oulu, Oulu, Finland
e-mail: [email protected]
M. Petrovic
Section of Geriatrics, Department of Internal Medicine and Paediatrics, Ghent University,
Ghent, Belgium
e-mail: [email protected]
A. Benetos
Department of Geriatrics, University Hospital of Nancy, FHU-CARTAGE Profiles and
INSERM DCAC, Université de Lorraine, Vandeoeuvre-les-Nancy, France
e-mail: [email protected]
<115/75 mmHg—a finding dating back long before the development of antihyper-
tensive drugs [5]—this would make practically all older people candidates for anti-
hypertensive treatment. On the other hand, a repeating finding in unselected
epidemiological cohorts is that low blood pressure is often associated with worse
prognosis [6–10]. Interestingly, this negative association between SBP and morbid-
ity/mortality was observed mainly in very frail subjects receiving antihypertensive
treatment and not in subjects without blood pressure lowering drugs [11–14]. How
should all this information be taken into account when assessing treatment of blood
pressure in an older patient?
Observational studies cannot confirm cause and effect and may even involve reverse
causality. In other words, frailty, subclinical disease, and low cardiac output may
lower blood pressure and simultaneously worsen prognosis leading to a biased rela-
tionship between high blood pressure and better prognosis. Therefore, it is urgent to
include in future therapeutic trials very frail older patients, i.e., those who usually
have been excluded from clinical trials in the past. Presently, we have evidence for
the relatively robust older patients (i.e., those with the “HYVET or SPRINT pro-
file”) in which our strategy should be very close to that proposed for the “younger-
old” subjects, i.e., 65–75 years old community-dwelling subjects. European
guidelines propose targeting BP levels <140/90 in these populations though for very
old frail subjects, such targets should be individualized. A treatment algorithm and
goals in various situations are presented in Fig. 18.1.
More generally, when antihypertensive treatment has been started earlier in life
and continuing without problems, the common sense indicates that the treatment
must not be modified or stopped due to age alone. As a whole, antihypertensive
treatment should be started clearly earlier, i.e., in midlife, to reach the full benefit of
treatment during the life course [15]. Good adherence and sufficient control of
blood pressure are essential also in an older patient, and prudent lifestyle advice
about diet and physical activity are useful. If problems, such as orthostatic
Preserved autonomy and function Preserved activities of daily living (ADL) but Loss of function and loss of
status Clinical Frailty Scale(CFS 1-3) reduced function (CFS4-6) autonomy for ADL; possibly with
limited life expectancy (CFS7-9)
Treatment outline and goal similar to Complete geriatric assessment in order to Consider antihypertensive treatment
younger adults individualize antihypertensive treatment from a holistic perspective
-If treated, start with one drug and
-Systolic blood pressure < 120-140 mm Only moderately altered Significantly altered proceed cautiously with systolic blood
Hg function: function: pressure goal < 150 mm Hg
-Start low and go slow to meet goal proceed according to proceed according to -Avoid using more than 3
-Always check supine and standing left panel right panel antihypertensive drugs, if possible
blood pressure to identify orthostatic
-Consider deprescribing
reaction
-Optimizeglobal cardiovascular -Identify other factors/drugs affecting
prevention blood pressure
Fig. 18.1 A treatment algorithm and goals in various situations of older patients with hypertension
18 Hypertension 231
burden for the heart is reduced. Too low diastolic may be harmful for brain circula-
tion [32, 33] and vulnerable patients must be carefully monitored.
It is important to exclude secondary hypertension also in the older patient, and the
diagnostic procedures are similar to those in younger patients. Especially in an older
patient, it is mandatory to measure blood pressure in both supine and standing posi-
tion in order to detect OH predisposing to falls. It is recommended that a 3-min
orthostatic test is always performed before medication changes. Isolated office
(“white coat”) hypertension is relatively common in an older patient, and its detec-
tion may require ambulatory measurement. On the other hand, office and its coun-
terpart isolated masked (home) hypertension are not innocuous phenomena [34] and
may indicate treatment, or at least careful follow-up. Pseudohypertension refers to
a situation when brachial blood pressure measurement gives a high reading due to
the pronounced arteriosclerosis. However, pseudohypertension is not so common,
and one should pose this diagnosis very carefully in order to avoid underestimation
of the importance of very elevated SBP levels in high-risk populations such as
patients with diabetes or severe renal failure. The condition may be a reason for
“resistant” hypertension and should be remembered, although confirmation may be
difficult without direct arterial measurement.
18.3.1 Lifestyle
There are surrogate data of the effects of lifestyle modification and hypertension in
old age. Prudent advice on a healthy diet, especially salt intake, and exercise are
recommended. Substituting ordinary salt to mineral salt is beneficial [35]. For
obese, weight reduction could lower blood pressure [36], but trial evidence is non-
existent for people 75 years and older, and malnutrition and development of sarco-
penia are possible hazards. Physical activity adjusted to clinical condition is always
recommended to sustain general health and functional capacity, but again without
specific trial evidence for the treatment of hypertension in an older patient. In gen-
eral, when treatment for hypertension is indicated, lifestyle modification alone is
seldom sufficient.
18 Hypertension 233
There are no specific differences in the selection of drug treatment between younger
and older patients, although it is often best to start low and go slow [26]. The main
drug classes of antihypertensives, i.e., diuretics, angiotensin-converting enzyme
inhibitors, angiotensin II receptor blockers, and calcium channel blockers, can be
used and with doses ultimately adjusted individually to reach appropriate targets. As
in younger patients, beta blockers are not recommended as first-line treatment with-
out specific indications. An interesting new possibility are the SGLT2-inhibitors,
which were initially developed as antidiabetic drugs. They lower blood pressure,
prevented CVD endpoints in RCTs, and are effective for heart failure (both systolic
and diastolic) even in nondiabetic patients [37, 38]. Experience of SGLT2-inhibitors
is accumulating also in older patients. Other drugs affecting blood pressure like
nitrates, alpha-blockers, and various psychotropic drugs must be remembered, espe-
cially as they may be prescribed by other specialists than general practitioners or
geriatricians. Although psychotropic drugs may not directly affect blood pressure,
they can nevertheless make a frail patient more susceptible to falls.
The general principle should be to start with monotherapy when treating older
patients especially frail patients over 80 years old. However, in most subjects, one
single BP-lowering drug is not enough to control BP, and therefore, patients and
their caregivers should be informed from the beginning about the possible adapta-
tion after the initiation of the treatment. Combining low doses of drugs with differ-
ent mode of action is usually better than increasing the dose of a single drug.
Dividing doses for morning and evening may be indicated, and there is ongoing
debate about the potential benefits of evening dosing [39]. Although maximum of
three drugs are sometimes recommended, this cannot be absolute, and treatment
must be individualized also for this.
Potential adverse effects and special considerations when using antihyperten-
sives for older patients are listed in Table 18.1.
A special case, not uncommon in an older patient with hypertension, is the acute
or subacute rise of blood pressure to very high levels (“hypertensive crisis”) [40,
41]. It is defined as systolic and diastolic blood pressure of >200 mmHg and
>130 mmHg, respectively, and divided to an emergency (signs of end-organ dam-
age) or urgency state without specific symptoms except blood pressure rise.
Emergency requires immediate treatment in a hospital setting and intensive care
unit, whereas urgency state can usually be treated ambulatory by enhancing oral
antihypertensive treatment with close follow-up. In both states, it is important to
avoid exaggerated treatment and reduce blood pressure safely.
234 T. E. Strandberg et al.
References
1. The Blood Pressure Lowering Treatment Trialists’ Collaboration. Age-stratified and blood
pressure-stratified effects of blood pressure-lowering pharmacotherapy for the prevention of
cardiovascular disease and death: an individual participant-level data meta-analysis. Lancet.
2021;398:1053–64.
2. Hughes D, Judge C, Murphy R, et al. Association of blood pressure lowering with
incident dementia or cognitive impairment: a systematic review and meta-analysis.
JAMA. 2020;323:1934–44.
3. SPRINT MIND Investigators for the SPRINT Research Group, Williamson JD, Pajewski NM,
Auchus AP, et al. Effect of intensive vs standard blood pressure control on probable dementia:
a randomized clinical trial. JAMA. 2019;321:553–61.
4. Strandberg TE, Benetos A, Petrovic M. Editorial: incident dementia in trials of antihyperten-
sive treatments. J Nutr Health Aging. 2019;23:914–5.
5. Strandberg TE. Blood pressure in a 100-year perspective. Circulation. 2019;140:101–2.
6. Boshuizen HC, Izaks GJ, van Buuren S, et al. Blood pressure and mortality in elderly people
aged 85 and older: community-based study. BMJ. 1998;316:1780–4.
7. Hakala SM, Tilvis RS, Strandberg TE. Blood pressure and mortality in an older population. A
5-year follow-up of the Helsinki Ageing Study. Eur Heart J. 1997;18:1019–23.
236 T. E. Strandberg et al.
8. Rastas S, Pirttilä T, Viramo P, et al. Association between blood pressure and survival over 9
years in a general population aged 85 and older. J Am Geriatr Soc. 2006;54:912–8.
9. van Bemmel T, Gussekloo J, Westendorp RG, et al. In a population-based prospective study,
no association between high blood pressure and mortality after age 85 years. J Hypertens.
2006;24:287–92.
10. van Bemmel T, Holman ER, Gussekloo J, et al. Low blood pressure in the very old, a conse-
quence of imminent heart failure: the Leiden 85-plus study. J Hum Hypertens. 2009;23:27–32.
11. Aparicio LS, Thijs L, Boggia J et al, International Database on Home Blood Pressure in
Relation to Cardiovascular Outcome (IDHOCO) Investigators. Defining thresholds for home
blood pressure monitoring in octogenarians. Hypertension. 2015;66:865–873.
12. Benetos A, Labat C, Rossignol P, et al. Treatment with multiple blood pressure medications,
achieved blood pressure, and mortality in older nursing home residents: the PARTAGE study.
JAMA Intern Med. 2015;175:989–95.
13. Mossello E, Pieraccioli M, Nesti N, et al. Effects of low blood pressure in cognitively impaired
elderly patients treated with antihypertensive drugs. JAMA Intern Med. 2015;175:578–85.
14. Streit S, Poortvliet RKE, Gussekloo J. Lower blood pressure during antihypertensive treat-
ment is associated with higher all-cause mortality and accelerated cognitive decline in the
oldest-old-data from the Leiden 85-plus study. Age Ageing. 2018;47(4):545–50. https://doi.
org/10.1093/ageing/afy072.
15. Suvila K, Niiranen TJ. Age of hypertension onset: potential for improving risk estimation and
hypertension management? Hypertension. 2021;78:1475–7.
16. Lipsitz LA. A 91-year-old woman with difficult-to-control hypertension a clinical review.
JAMA. 2013;310:1274–80.
17. Masuo K, Mikami H, Ogihara T, et al. Changes in frequency of orthostatic hypotension in
elderly hypertensive patients under medications. Am J Hypertens. 1996;9:263–8.
18. Juraschek SP, Taylor AA, Wright JT Jr. Orthostatic hypotension, cardiovascular outcomes, and
adverse events: results from SPRINT. Hypertension. 2020;75:660–7.
19. Sheppard JP, Burt J, Lown M et al.; OPTIMISE Investigators. Effect of antihyperten-
sive medication reduction vs usual care on short-term blood pressure control in patients
with hypertension aged 80 years and older: the OPTIMISE randomized clinical trial.
JAMA. 2020;323:2039–2051.
20. Sheppard JP, Lown M, Burt J, et al. Blood pressure changes following antihypertensive medi-
cation reduction, by drug class and dose chosen for withdrawal: exploratory analysis of data
from the OPTIMISE trial. Front Pharmacol. 2021;12:619088.
21. Applegate WB, Williamson JD, Berlowitz D. Deprescribing antihypertensive medication in
elderly adults. JAMA. 2020;324:1682.
22. Beckett NS, Peters R, Fletcher AE, et al. Treatment of hypertension in patients 80 years of age
or older. N Engl J Med. 2008;358:1887–98.
23. SPRINT Research Group, Wright JT Jr, Williamson JD, et al. A randomized trial of intensive
versus standard blood-pressure control. N Engl J Med. 2015;373:2103–16.
24. SPRINT Research Group, Lewis CE, Fine LJ, Beddhu S, et al. Final report of a trial of inten-
sive versus standard blood-pressure control. N Engl J Med. 2021;384:1921–30.
25. Williamson JD, Supiano MA, Applegate WB et al.; SPRINT Research Group. Intensive vs
standard blood pressure control and cardiovascular disease outcomes in adults aged ≥75 years:
a randomized clinical trial. JAMA. 2016;315:2673–2682.
26. Benetos A, Rossignol P, Cherubini A, Joly L, Grodzicki T, Rajkumar C, Strandberg TE,
Petrovic M. Polypharmacy in the aging patient: management of hypertension in octogenar-
ians. JAMA. 2015;314:170–80.
27. Benetos A, Petrovic M, Strandberg T. Hypertension management in older and frail older
patients. Circ Res. 2019;124:1045–60.
28. Warwick J, Falaschetti E, Rockwood K, et al. No evidence that frailty modifies the positive
impact of antihypertensive treatment in very elderly people: an investigation of the impact of
frailty upon treatment effect in the HYpertension in the Very Elderly Trial (HYVET) study, a
18 Hypertension 237
19.1 Introduction
T. L. De Backer (*)
Cardiovascular Department, Ghent University Hospital, Ghent, Belgium
Clinical Pharmacology, Ghent University, Ghent, Belgium
e-mail: [email protected]
A. A. Mangoni
Discipline of Clinical Pharmacology, Flinders University and Flinders Medical Centre,
Adelaide, South Australia
e-mail: [email protected]
including primary and secondary healthcare staff as well as with the patient and/or
her/his caregivers (Figs. 19.1 and 19.2).
Optimising pharmacotherapy in older patients with heart failure is discussed.
Device-based treatments (cardiac resynchronisation therapy, implantable cardio-
verter defibrillator, assist devices) and heart transplantation considered for selected
advanced heart failure patients do not fall within this scope.
Caregivers at home
or nursing home
Multidiscipinary team
GP, geriatrician, Patient
cardiologist, surgeon, Old & Relatives, Friends,
nurse, pharmacologist, Heart failure Neighbours
social worker,
physiotherapist,
psychologist, dietician
Health care
system
Fig. 19.1 Parties involved in the care of the old heart failure patient
19 Heart Failure 241
Risk Factors
Polypharmacy
Patient Risk Modifiers
intracardiac pressures and/or inadequate cardiac output at rest and/or during exer-
cise [3].
Although definitions and interpretations may vary and may be somewhat artifi-
cial, the definition of left ventricular HF currently includes three phenotypes based
on left ventricular ejection fraction: heart failure with preserved ejection fraction
(HFpEF, LVEF ≥50%, with evidence of structural and/or functional cardiac abnor-
malities and/or raised natriuretic peptides (NPs)), mildly reduced (HFmrEF, LVEF
41–49%, mildly reduced LV systolic function), and reduced ejection fraction
(HFrEF, LVEF <40%, significant reduction in LV systolic function) [3]. Heart fail-
ure can also be due to right ventricular dysfunction assessed by RV function mea-
surements. Right ventricular heart failure is a syndrome by its own, however often
due to and in association with LV heart failure.
There are many potential causes of heart failure having an ischaemic and/or non-
ischaemic origin. Of note, in the ageing population, an increased prevalence and
incidence of senile cardiac amyloidosis or wild-type transthyretin amyloidosis
(ATTRwt) often in combination with aortic valve stenosis are noted.
Although prognosis of patients with HF, mainly HFrEF, has improved since the
publication and emergence on the market of successful HF treatment trials, progno-
sis remains poor, and quality of life (QOL) markedly reduced.
Mortality rates are higher in observational studies than in clinical trials [11]. In
the Olmsted County cohort, 1-year and 5-year mortality rates after diagnosis, for all
types of HF patients, were 20% and 53% [12]. In a report combining results of the
Framingham Heart Study (FHS) and Cardiovascular Health Study (CHS) cohorts,
mortality rate within 5 years following diagnosis was 67% [13].
A better overall survival in HFmrEF and HfpEF compared to HfrEF was consid-
ered. The MAGGIC meta-analysis concluded that the adjusted mortality risk in
patients with HfpEF was lower than in patients with HfrEF. However, most obser-
vational studies show that this difference is negligible [12, 13]. In the Olmsted
county cohort of hospitalised patients, it was shown that the prevalence of HFpEF
over 15 years increased together with an increase in hypertension, atrial fibrillation,
and diabetes and that the death rate remained unchanged (29% at 1 years and 65%
at 5 years) which was minimally lower than the death rate for HFrEF (32% and
68%, respectively) [14].
The various and evolving definitions and categorisations of the heart failure syn-
drome over time together with heterogeneity in patient characteristics and the evo-
lution in prevention and treatment also may contribute to the varying numbers in
epidemiological data. Data from GWTG-HF (Get With The Guidelines–Heart
Failure) linked to Medicare data looking at 5-year outcomes and median survival in
patients hospitalised for heart failure showed a median survival of 2.1 years and a
similar high 5-year mortality, 75%, in HFrEF, HFbEF(borderline, =mrEF), and
HFpEF. Also, the composite of mortality and rehospitalisation was similar for all
subgroups, with cardiovascular and HF readmission rates being higher in HFrEF
and HFbEF compared to HFpEF. Conclusion was that patients with HF across all
ages and EF groups had a markedly lower median survival compared to the overall
population [15, 16].
Age by itself is an independent risk factor for mortality. Patients >75 years have
significantly higher in-hospital mortality rates than patients <75 years (11.2% vs
4.4%) (www.nicor.org.uk). Two nationwide cohorts of patients hospitalised for
new-onset heart failure based on linkage of the Dutch Hospital Discharge Registry
and the National Cause of Death registry assessed overall and cause-specific mortal-
ity rates over time stratified by age and sex. Mortality rates at 1 year were high and
increased considerably with age similarly in men and women, from 10.5% in
women aged 25–54 years to 46.1% in women aged >85 years and from 9.8% in men
aged 25–54 years to 53.4% in men >85 years, although the overall 1-year absolute
risk of death across all ages declined by 4.0% in men and 3.2% in women [17]. The
Cardiovascular Health Study, a longitudinal cohort study of community-dwelling
older adults >65 years in the USA, reported 1-, 5-, and 10-year mortality rates of
19%, 56%, and 83%, respectively, following the onset of HF [13]. The Canadian
Chronic Disease Surveillance System data confirmed that once HF develops, mor-
tality increases exponentially with age [18]. Hospital admissions and readmissions
are frequent in heart failure patients for cardiac as well as non-cardiac reasons and
negatively impact prognosis. The proportion of hospitalisations related to
244 T. L. De Backer and A. A. Mangoni
Comprehensive
Geriatric Assessment
Mainstay Pharmacotherapy
ARNI or ACEI (ARB)
b-blocker
MRA
SGLT-2inhibitor
Loop Diuretics
Cardiac rehabilitation,
Consider
Education &
Self-management, Digoxin
Weight, Fluid & Food Hydralazine/ISSDN Devices,
intake, Medication Ivabradine percutaneous
Reviews, intervention,
Iron iv
Telemonitoring, surgery
Life Care Plan
The ageing population is growing, heart failure increases with age, and age is an
independent predictor of morbidity and mortality. With a few exceptions, hard evi-
dence on the best treatment for heart failure specifically in older persons is lacking
since they were largely excluded from major HF trials [22]. In the earlier studies
over the past 40 years, the upper limit of age was often put at patients of 65 up to
70–75 years old and with little or no comorbidities. The more recent trials also
allowed patients >75 years old, being appropriate and logical given the growing
older population. However, evidence in the very old and/or multimorbid and/or frail
older person is virtually non-existent.
Most drug trials until now provided evidence for a positive benefit-risk effect on
morbidity and mortality mainly in HFrEF, while almost none could prove signifi-
cant benefit on hard outcomes as mortality in HFpEF. Few trials demonstrated
symptomatic benefit and reduced hospitalisations in HFpEF.
The recent EMPEROR-preserved and DELIVER trials showed a significant
reduction by empagliflozin and dapagliflozin in the combined endpoint of CV death
and HF hospitalisations, driven by a reduction in hospitalisation [23]. Treatment of
HFpEF is quite individualised and mainly involves treatment of risk factors (such as
hypertension) and comorbidities (such as atrial fibrillation, coronary artery disease,
diabetes, renal insufficiency, pulmonary hypertension, thyroid disorder, etc.) and
control of symptoms (fluid balance).
To deal with age, most trials specified subgroup analyses looking at different age
ranges or at older versus younger participants, often defined as <65 years and
>65 years old. This was done for most trials with ACE inhibitors (ACE-I), beta-
blockers (BB), angiotensin receptor blockers (ARB), digoxin, hydralazine/ISDN,
aldosterone antagonists/mineralocorticoid receptor antagonist (MRA), ivabradine,
angiotensin receptor-neprilysin inhibitors (ARNI), and sodium-glucose cotrans-
porter 2 inhibitors (SGLT2i).
An increasing number of post hoc analyses of the effectiveness of HF treatment
in a broader range of older patients are performed.
A BIOSTAT-CHF analysis assessing the association between up-titration of
angiotensin-converting enzyme inhibitors (ACEI), angiotensin receptor blockers
(ARB), and beta-blockers and outcome across the age spectrum in HFrEF patients
showed that achieving higher doses of ACEI/ARB was associated with improved
outcome regardless of age, while achieving higher doses of beta-blockers was asso-
ciated with improved outcome only in younger but not in older patients. The incon-
sistent association between beta-blocker dosage and heart rate in older patients is
hypothesised to be due to a decreased cardiovascular responsiveness to beta-
adrenergic stimulation, already acting as an intrinsic beta-blockade, as reflected by
the lower resting heart rate in older patients [24]. Heart rate reduction might be
more important than the achieved beta-blocker dose in HFrEF. For example, in
CIBIS-ELD, the achieved heart rate target rather than beta-blocker dose (carvedilol
or bisoprolol) was significantly associated with outcome (see further, [25, 26]).
246 T. L. De Backer and A. A. Mangoni
Also, the higher prevalence of atrial fibrillation in older patients might lower the
beneficial effects of beta-blockers.
One systematic review and meta-analysis of the efficacy (mortality and hospitali-
sations for cardiovascular causes) and safety of MRA treatment in older HF patients
≥65 years reported overall benefit from MRA therapy with no significant treatment
interaction for age in patients with HFrEF, while no significant effect on any effi-
cacy outcome in patients with HFpEF. In a subanalysis in patients ≥75 years with
HErEF, the effect of MRA treatment on overall mortality seemed uncertain, mainly
due to heterogeneity [25].
In an IPD meta-analysis of RALES, EMPHASIS-HF, and TOPCAT in patients
≥75 years versus <75 years of age, the primary outcome (composite of death from
cardiovascular causes or hospitalisation for HF), cardiovascular death, and all-cause
death were reduced by MRAs without significant between-trial or age (younger vs.
older) heterogeneity. Worsening renal function (without more hyperkalaemia) was
found more frequently in older patients. The reduction in morbidity and mortality
was more marked in patients with HFrEF however overall homogenous across
HFrEF and HFpEF (in HFpEF lower events, but not statistically significant). The
authors pleaded for measures to increase MRA treatment in the older popula-
tion [26].
A cohort study using US Medicare fee-for-service claims data (2014–2017) in
patients >65 years with HFrEF evaluating the effectiveness of angiotensin receptor-
neprilysin inhibitor (ARNI) versus renin-angiotensin system inhibition (RAS)
found that ARNI treatment was associated with lower risk of the composite effec-
tiveness endpoint (worsening HF or all-cause mortality) compared with RAS block-
ade alone [27].
In the PARADIGM-HF trial (LCZ696 vs. enalapril), efficacy (CV death or heart
failure hospitalisation) and safety according to age (<55 years (n = 1624),
55–64 years (n = 2655), 65–74 years (n = 2557), and ≥75 years (n = 1563)) were
consistent across all age categories with LCZ696 having a favourable benefit–risk
profile in all age groups [28].
A meta-analysis of the effects of the SGLT2 inhibitors dapagliflozin (DAPA-HF)
and empagliflozin (EMPEROR-reduced) on cardiovascular outcomes in patients
with HFrEF with or without diabetes showed a significant reduction in all-cause
death (−13%), cardiovascular death (−14%), combined endpoint of cardiovascular
death or first hospitalisation for heart failure (−24%), composite endpoint of recur-
rent hospitalisations for heart failure or cardiovascular death (−25%), and compos-
ite endpoint of renal events (−38%). Analysis based on age groups, < and >65 years,
showed a persistent 25% reduction in the composite endpoint of cardiovascular
death or hospitalisation for heart failure [29].
Empagliflozin in patients with HFpEF (in the EMPEROR-Preserved trial defined
as EF >40%) and with or without diabetes lowered the composite of cardiovascular
death or hospitalisation for heart failure by 21%, mainly due to lower risk of hospi-
talisation for heart failure (29% reduction). Based on age, the endpoint reduction in
patients <70 years was −12% (0.88, 0.70–1.11), while it was −25% (0.75, 0.64–0.87)
in patients >70 years. If confirmed in other trials (confirmed for dapagliflozin in
19 Heart Failure 247
DELIVER trial) and in real-world data, SGLT2 inhibitors are the first class showing
significant beneficial effect in HFpEF (subgroup analysis showed significance in EF
>40 to <50% and >50% to <60%, in EF >60% NS) [23].
Pooled data on the safety (12 studies) and efficacy (9 studies; outcome: HbA1c
reduction) of the SGLT2 inhibitor dapagliflozin 10 mg/day monotherapy or com-
bined with oral antidiabetic drugs or with insulin in older patients >65 years (20%
of the studied populations) with type 2 diabetes showed HbA1c reductions across
age subgroups (<65 years and >65 years) with greater reductions in HbA1c in
patients aged <65 years than in patients aged >65 years. Hypoglycaemia, volume
depletion, and renal AEs (predefined serum creatinine thresholds) occurred more
often in patients aged >65 years, reflecting the lower eGFR in the older persons.
Orthostatic hypotension occurred in <0.1% in both age groups. Urinary tract and
genital infections were similar for both DAPA-treated age groups. It was concluded
that DAPA’s efficacy and safety observations indicate a practical, new approach to
the treatment of T2DM patients aged >65 years [30].
Very few trials investigated the efficacy and safety of drug therapy specifically in
an older population.
The PEP-CHF trial in patients >70 years of age and with evidence of diastolic
dysfunction, comparing placebo with perindopril 4 mg/day, showed a clear trend
towards reduction in the primary endpoint of combined all-cause mortality and
heart failure hospitalisation (HR 0.692: 95% CI 0.474–1.010; P = 0.055), however
not reaching statistical significance due to insufficient power. A significant reduc-
tion in hospitalisation for heart failure and a significant improvement in functional
class and 6-min walking distance at 1 year were observed in patients receiving per-
indopril [31].
The SENIORS trial assessed the effect of nebivolol, a beta-blocker with vasodi-
lating properties, on mortality and cardiovascular hospital admission in older
patients >70 years with heart failure, regardless of ejection fraction, compared to
placebo. The primary outcome, a composite of all-cause mortality or cardiovascular
hospital admission, was reduced by 34%(HR, 0.86; 95% CI, 0.74–0.99; P = 0.039),
with no significant influence of age, gender, or ejection fraction on the effect of
nebivolol on the primary outcome. For all-cause mortality HR was 0.88 (95% CI,
0.71–1.08; P = 0.21). Nebivolol was considered an effective and well-tolerated
treatment for heart failure in the older population [32].
As part of the larger CIBIS trial, the CIBIS–ELD trial was a tolerability superior-
ity study of bisoprolol versus carvedilol comparing the tolerability and clinical
effects of the two beta-blockers in older patients with heart failure with reduced or
preserved left ventricular ejection fraction. The primary endpoint tolerability,
defined as reaching and maintaining guideline-recommended target doses after
12 weeks treatment, was comparable, 24% versus 25%. Bisoprolol treatment
resulted in greater reduction of heart rate (adjusted mean difference, 2.1 bpm; 95%
CI, 0.5–3.6; P = 0.008) and more, dose-limiting, bradycardic adverse events (16 vs.
11%; P = 0.02), while carvedilol intake resulted in a reduction of forced expiratory
volume (adjusted mean difference, 50 mL; 95% CI, 4–95; P = 0.03) and more, non-
dose-limiting, pulmonary adverse events (10 vs. 4%; P = 0.001). It was concluded
248 T. L. De Backer and A. A. Mangoni
that overall tolerability to target doses was comparable, while the pattern of intoler-
ance was different with bradycardia occurring more often in the bisoprolol group,
whereas pulmonary adverse events occurring more often in the carvedilol group [33].
A 4-year follow-up study of the CIBIS-ELD trial investigated the prognostic
value of achieved heart rate after beta-blocker optimisation on long-term mortality.
Heart rate increases by 10 bpm following beta-blocker up-titration which was asso-
ciated with a subsequent mortality hazard ratio of 1.19 (95% CI, 1.02–1.38;
P = 0.023). The heart rate range with the lowest mortality and the fewest treatment-
related adverse events was 55–64 bpm. The achieved beta-blocker dose was not
associated with mortality risk. It was concluded that the achieved heart rate after
up-titration, but not the beta-blocker dose, predicted all-cause mortality risk in older
patients with chronic
heart failure and that titration should aim for resting heart rates between 55 and
64 bpm [34].
Several smaller, mainly Japanese, studies looked at real-world data on the effec-
tiveness and safety of SGLT2 inhibition in older persons.
The STELLA-ELDER study was a post-marketing surveillance (PMS) study inves-
tigating real-world evidence on the safety of ipragliflozin in older (>65 years) Japanese
patients with type 2 diabetes mellitus. Of all patients, 16.91% experienced 1880 adverse
drug reactions (ADR), of which 165 ADRs were considered serious in 1.49% of the
patients. The most common ADRs were skin and subcutaneous tissue disorders (3.25%),
renal and urinary disorders (2.74%), and infections (2.23%). Of special interest were
skin complications, volume depletion, polyuria/pollakisuria, genital and urinary tract
infection, renal disorders, hypoglycaemia, cerebrovascular disease, cardiovascular dis-
ease, malignant tumour, fracture, and ketone body-related events. Risk factors identified
for hypoglycaemia were BMI (<18.5 kg/m2 vs. ≥22.0 to <25.0 kg/m2; OR, 9.356) and
concomitant insulin use (vs. no; OR, 4.946), and risk factors identified for volume
depletion were age (≥75 years vs. <75 years; OR, 1.737) and concomitant loop diuretic
use (vs. no; OR, 2.105). Overall, there were no safety concerns that were not previously
observed in pre-marketing trials. The conclusion was that this PMS revealed probable
ADRs in older Japanese patients with T2DM taking ipragliflozin with no new safety
concerns and that PMS should help physicians to identify possible treatment-emergent
ADRs in ipragliflozin-treated patients in real-world settings [35].
Another Japanese study assessing the use of sodium-glucose cotransporter 2
inhibitors in older patients (mean age 73.7 ± 10 years) with type 2 diabetes mellitus
did not report events associated with oral administration of SGLT-2 inhibitors after
a mean follow-up period of 289.3 days. Fasting blood glucose and glycosylated
haemoglobin levels at 6 months were significantly lowered without signs of intra-
vascular collapse or changes in creatinine, blood urea nitrogen/creatinine ratio, or
estimated glomerular filtration rate. Treatment with SGLT-2 inhibitors did signifi-
cantly increase haemoglobin and haematocrit levels. In a small subgroup with a
brain natriuretic peptide level exceeding the normal upper limit before treatment
with SGLT-2 inhibitors, the brain natriuretic peptide levels significantly decreased
after 6 months of treatment. SGLT-2 inhibitors at 6 months exerted a favourable
hypoglycaemic effect and no signs of dehydration were observed [36].
19 Heart Failure 249
hospitalisation for heart failure, and 25% reduction in the composite of cardiovas-
cular death or recurrent hospitalisations for heart failure. For patients with HFpEF
with or without diabetes, empagliflozin reduced the composite of cardiovascular
death or hospitalisation for heart failure by 21% in the overall population and 25%
in patients >70 years. If confirmed in other trials and in real-world data, SGLT 2
inhibitors are the first class showing beneficial effect in HFpEF. As a comparison, in
earlier trials, the ARB candesartan, MRA spironolactone, and Sacubitril-Valsartan
also showed some modest effect on CV death and HF hospitalisation (up to 15%
risk reduction) in HfpEF patients, however mainly in the groups with an EF 40–49%
considered as mildly reduced EF [23].
Overall, it can be assumed that older patients also benefit from these evidence-
based pharmacological treatments and that guideline recommended HF treatment
aiming at improving the quality of patient care should also be applied in older peo-
ple. A guideline gives an advice, not an order, and a benefit-risk balance should be
made for each individual patient.
Current mainstay drug therapy recommended for all heart failure patients are
ARNI or ACE-I if ARNI not tolerated (ARB only if ARNI and ACE-I not tolerated),
beta-blockers, MRA, and SGLT2i as they improve prognosis by reduction of mor-
bidity and mortality. Dosage should be started low and slow and then progressively
increased based on tolerance. Symptomatic treatment with loop diuretics and acet-
azolamide in certain cases (ADVOR trial) is given in case of congestion to improve
symptoms, exercise tolerance, and reduce HF hospitalisations.
The following drugs should or might be considered in specific conditions.
Ivabradine, an If channel blocker, should be considered in case of EF <35%, sinus
rhythm with a heart rate >70 bpm in patients on mainstay HF therapy (with or with-
out a beta-blocker). Digoxin might be considered for symptomatic patients on base-
line HF therapy. Hydralazine/isosorbide dinitrate might be considered in
symptomatic patients not tolerating any of the baseline therapies.
Some other drug classes for use in HF are being investigated; however, current
evidence is limited. The soluble guanylate cyclase stimulator vericiguat might be
considered in patients with HFrEF, on top of standard therapy to further reduce the
risk of CV mortality and hospitalisations for HF (VICTORIA study) [40]. The car-
diac myosin activator omecamtiv mecarbil might be considered for HFrEF patients
on top of standard therapy to reduce the risk of CV mortality and HF hospitalisation
(GALACTIC-HF study) [41–44].
Besides optimal HF drug therapy, other cardiovascular and non-CV therapies,
including non-pharmacological strategies, should be applied in the setting of cardio-
vascular and non-cardiovascular risk factors and comorbidities, which may play a
role in initiating and worsening of heart failure directly and/or indirectly (Fig. 19.3).
In older patients, heart failure seldom comes alone. Several factors may play a role
in the initiation and/or worsening of heart failure, may compromise or hamper opti-
mal therapy, and may as such influence prognosis. The list of risk factors and
19 Heart Failure 251
Especially older patients with HF have increased risk for malnutrition and deficien-
cies, which should be checked for and treated accordingly [47]. In the OPTIMIZE-HF
study, low sodium serum levels was a predictor of in-hospital mortality [48].
Psychosocial stress (mental, financial, familial, loneliness, anxiety, etc.) increases
cardiovascular risk and should be assessed (e.g., via Patient Health Questionnaire),
and psychosocial support should be given when necessary. Increased attention and
support in patients with mental disorders and cardiovascular disease is recom-
mended to help improve adherence to lifestyle changes and medication.
Blood pressure should be regularly measured, and hypertension, an important
risk factor in heart failure, should be prevented and/or efficiently treated with life-
style interventions (no smoking, salt intake <5 g/day, physical exercise, weight con-
trol, stress reduction) and antihypertensive drugs (main classes: RAS inhibitors,
diuretics, calcium channel blockers, beta-blockers). Blood pressure should also be
measured in the standing position and/or after a meal to detect orthostatic and/or
postprandial hypotension, especially in older persons. A blood pressure target of
<140/90 mmHg with ranges of SBP <140 to 130 and DBP <80 to 70 mmHg are
recommended, if tolerated. In older persons >80 years with good functionality, an
antihypertensive treatment as given to younger adults should be considered. In the
older persons with a moderate functional limitation and preserved autonomy for the
activities of daily living, a geriatric risk assessment might be performed to decide on
the individual treatment approach. Patients with few comorbidities and only very
limited loss of autonomy can be considered to receive the same antihypertensive
regimen as the older persons with good functionality. In older persons with severe
functional limitations and loss of autonomy and/or very reduced life expectancy,
quality of life and symptom control is the main aim, and treatment should be regu-
larly reviewed and discussed in a multidisciplinary context. Because of safety rea-
sons, treatment might be reduced or stopped [49–52].
Hypercholesterolemia should be treated with lifestyle measures (physical activ-
ity, healthy diet) and lipid-lowering drugs (mainly statins). The targets depend on
the overall cardiovascular risk and tolerance. In calculating cardiovascular risk in
older persons, often important non-CVD risk has to be taken into account, and
therefore the Older Person SCORE and LIFE-CVD scores are more appropriate.
Guidelines recommend statins in older people (>65 years) with atherosclerotic car-
diovascular disease in the same way as in younger people. In primary prevention,
according to level of risk, statins are recommended in older people <75 years. In
people >75 years, initiation of statins may be considered if at high risk. In older
patients, one should be alert for myopathy. Although there is less direct evidence of
benefits in the primary prevention setting among patients older than 75 years, results
from RCTs support the use of statins in older patients at high or very high risk of
ASCVD. The results from the STAREE (Statin Therapy for Reducing Events in the
Elderly; NCT02099123), from the SITE/SAGA (Statins In The Elderly;
NCT02547883), and PREVENTABLE (Pragmatic Evaluation of Events and
Benefits of Lipid-Lowering in Older Adults; NCT04262206) trials will be of help to
elucidate some of the questions that remain unanswered. Shared decision-making
on statin therapy in 75+-year-old people in primary prevention should be based on
19 Heart Failure 253
good clinical judgment, comorbidities, frailty, patient preferences, and life expec-
tancy [53].
Diabetes mellitus, considered more as a comorbidity, should be as much as pos-
sible prevented by lifestyle measures (weight control, physical activity). Once DM
is established, pharmacological treatment should be installed taking into account
age, risk factors, and comorbidities. SGLT2 inhibitors are the preferred choice in the
setting of CV disease and heart failure. Overall, in diabetes patients, an HbA1c
target of <7% is recommended. Based on personal physical and mental health sta-
tus, less rigorous HbA1c and glucose targets should be considered in older patients,
ranging from <7.5% in healthy older persons up to <8.5–9% in very complex older
patients with a poor overall health status [54, 55].
Atrial Fibrillation
The incidence and prevalence of atrial fibrillation (AF), the most frequent cardiac
arrhythmia worldwide, typically increases with age and with HF severity [56].
AF and HF frequently coexist and can cause or exacerbate each other through
several mechanisms such as structural cardiac remodelling, activation of neurohor-
monal systems, and heart rate-related LV impairment [57–60]. However, if AF is the
cause of HF (called tachycardiomyopathy), the clinical course may be more favour-
able than with other causes of HF [61]. On the contrary, if AF develops in patients
with chronic HF, prognosis seems worse, with increased stroke and increased mor-
tality [62, 63].
The management of patients with concomitant HF and AF includes identification
and treatment of potential causes and/or triggers of AF, management of HF, preven-
tion of thromboembolic events, rate control, and/or rhythm control.
Potential causes or precipitating factors, such as thyroid disorders, electrolyte
disturbances, infections, uncontrolled hypertension, and valve disease, should be
checked and corrected. Heart failure should be optimally managed with the main-
stay therapy, and increased congestion due to AF should be managed with diuretics.
Oral anticoagulants (OACs) are the cornerstone of treatment of AF to reduce the
risk for stroke/systemic thromboembolism, and—unless contraindicated—long-
term oral anticoagulant therapy is recommended in all patients with HF and parox-
ysmal, persistent, or permanent AF and CHA2DS2-VASc score >2 in men and >3 in
women. Four large randomised controlled trials (RCTs) have shown that non-
vitamin K antagonist oral anticoagulants (NOACs) also called direct oral anticoagu-
lants (DOACs) are non-inferior to vitamin K antagonists (VKAs) in preventing
stroke and systemic embolism, as well as regarding their risk for major bleeding.
Direct-acting oral anticoagulants (DOACs) are preferred for the prevention of
thromboembolic events in patients with AF—except in patients with moderate or
severe mitral valve stenosis and mechanical prosthetic heart valves—as they have
254 T. L. De Backer and A. A. Mangoni
Valve Disease
The prevalence of valve disease is increasing with age, with more than 10% of
people aged 75 years and older having moderate or severe valve disease.
Improved surgical techniques and percutaneous valve interventions increase the
treatment options for older patients with or without comorbidities.
Aortic valve stenosis and mitral valve and tricuspid valve regurgitation are com-
mon in older people. Aortic valve stenosis and mitral valve regurgitation are the two
most prevalent left-sided valve pathologies observed in older people.
Risk factors associated with the development of AS in older adults may be simi-
lar to those for atherosclerosis. In the Cardiovascular Health Study evaluating 5201
persons aged ≥65 years old, the overall incidence of aortic sclerosis and AS was
26% and 2% increasing up to 37% and 2.6% in the 75 years old. Major independent
clinical risk factors for degenerative aortic valve disease were age with a twofold
increased risk for each 10-year increase in age, male sex with a twofold excess risk,
current cigarette smoking with a 35% increased risk, and hypertension with a 20%
increased risk. High serum concentrations of lipoprotein(a) and low-density lipo-
protein cholesterol were also significant risk factors [77]. Aortic valve stenosis may
cause or worsen HF by increasing LV afterload with consequent LV hypertrophy
and remodelling [79].
Once heart failure symptoms (angina, dyspnoea, syncope) occur in patients with
severe aortic valve stenosis, the prognosis is very poor. There are no pharmacologi-
cal therapies specifically for improving outcomes of AS. However, HF patients with
symptomatic aortic valve stenosis should receive standard HF therapy, paying extra
attention to the risk of hypotension.
Aortic valve intervention is recommended in patients with HF symptoms and
severe, high-gradient aortic valve stenosis (valve area <1 cm2, mean gradient
>40 mmHg), regardless of LVEF and with a life expectancy of >1 year.
In low-gradient aortic valve stenosis, haemodynamic assessment (with dobuta-
mine) might be necessary to determine further management [80].
Older patients have higher in-hospital mortality than younger patients, due to a
poorer preoperative general status, more extensive age-related valve calcifications,
and fragile connective tissues, and if CABG, aneurysm repair, and mitral valve sur-
gery are concomitantly performed. Death after 30 days is more likely (up to 70%)
due to non-cardiac causes as pneumonia, stroke, and malignancy. However, older
patients who do survive the perioperative period, mostly do well with a functional
and quality of life improvement. In older patients referred for cardiac surgery, a
global assessment integrating frailty, disability, and risk scores should characterise
elderly patients better and identify those at increased risk [81].
Transcatheter aortic valve implantation (TAVI) is non-inferior to surgical aor-
tic valve replacement (SAVR) in reducing clinical events and mortality and dis-
abling stroke in patients at high and intermediate risk for surgery [82–84]. In
low-risk patients, SAVR is recommended in patients <75 years and at low surgical
risk (STS-PROM score or EurCORE II <4%), whereas TAVI is recommended in
patients >75 years or at high surgical risk (STS-PROM score or EuroSCORE II
>8%). In specific cases, balloon aortic valvuloplasty may be considered as in
highly symptomatic patients with acute heart failure and shock as a bridge to
TAVI or SAVR [80].
A multidisciplinary team (cardiologists, cardiac surgeons, geriatricians, nurses)
should make the benefit risk balance of TAVI and SAVR according to the patients
age, medical history, clinical features, anatomical aspects, life expectancy, and
patient preference.
19 Heart Failure 257
Mitral Regurgitation
Mitral regurgitation is a common valve abnormality in older adults with an esti-
mated odds ratio of 1.3 per 10-year increase in age.
Mitral regurgitation can be caused by organic or primary mitral regurgitation and
functional or secondary ischemic or non-ischaemic mitral regurgitation.
Organic or primary mitral regurgitation is due to intrinsic disease of the mitral
leaflets or subvalvular apparatus such as calcific degeneration, mitral valve pro-
lapse, flail mitral leaflet, and ruptured chordae tendineae. Surgery, preferably repair,
is recommended in patients with severe primary MR and HF symptoms.
Functional or secondary mitral regurgitation is mainly a disease of the left ven-
tricle. Functional or secondary ischemic mitral regurgitation is due to ischaemia or
papillary muscle infarction, with or without rupture, associated with an acute myo-
cardial infarction. Functional secondary non-ischaemic mitral regurgitation is due
to apical displacement of the papillary muscles associated with annular dilatation
and left atrial dilatation caused by left ventricular dilatation of any origin. Secondary
mitral regurgitation is a dynamic condition, and echocardiographic quantification
during exercise may be helpful in patients with moderate regurgitation at rest and
developing symptoms during physical activity [80, 85].
In older patients, the most common causes of mitral regurgitation referred for
mitral valve surgery are prolapse due to myxomatous degeneration and ischaemic
heart disease. Moderate or severe mitral regurgitation is associated with a poor
prognosis in patients with HF [86, 87]. Follow-up by a multidisciplinary heart team
is recommended. Optimal medical therapy, including cardiac resynchronisation
therapy, should be considered. In patients with severe mitral regurgitation and
HFrEF requiring revascularisation, mitral valve surgery and CABG should be con-
sidered. Isolated mitral valve surgery may be considered in symptomatic patients
with severe mitral regurgitation despite optimal therapy and at low surgical risk [80].
Chronic mitral regurgitation is the second most common reason for valve surgery
in older adults, after aortic valve surgery. While in younger asymptomatic patients
with early evidence of left ventricular dysfunction, surgery is recommended, in
patients over 80 years old, surgery might be considered only when symptoms are
present since postoperative outcomes are less favourable in this group. The risk
associated with the intervention, repair as well as with replacement, increases with
the presence of ischaemic heart disease, extent of calcification, and concomitant
interventions. Outcome after surgery in older patients varies with age and disease
severity and is better after mitral valve repair than replacement. However, late sur-
gery might contribute more than age to worse outcome in mitral valve surgery in
older adults <80 years.
Mitral valve repair, if feasible, is the surgical treatment of choice for mitral regur-
gitation and HF symptoms, also in older patients, as well as in primary MR and
secondary MR. If surgery is contraindicated or considered at high risk, then percu-
taneous repair may be considered [80, 88–90].
258 T. L. De Backer and A. A. Mangoni
19.6.3.1 Cancer
As heart failure increases with age, so does cancer. The incidence rates for cancer
overall go from fewer than 25 cases per 100,000 people in age groups under
20 years, to about 350 per 100,000 people in ages 45–49 years, to more than 1000
per 100,000 people in ages 60 years and older.
On the one hand, the risk of heart failure is increased in cancer patients deter-
mined by the cancer itself, cancer treatments, age, and underlying existing cardio-
vascular risk factors and comorbidities. Older people compared to younger people
seem more susceptible to cardiotoxicity related to cancer therapies. The cardiotoxic
effects of cancer therapies may be directly through cardiomyocyte damage or indi-
rectly through ischaemia, myocarditis, systemic arterial hypertension, pulmonary
hypertension, valvular heart disease, and arrhythmia.
On the other hand, the risk of worse cancer outcome in HF patients is increased
due to not starting or withdrawing anticancer therapies [98].
Management of HF in cancer patients should be done in a multidisciplinary
approach by preference in a cardio-oncology service and according to the stan-
dard guidelines of heart failure. Baseline cv risk assessment and a HFA-ICOS risk
assessment to define the risk for cardiotoxicity are advised [99]. Close clinical
and echocardiographic follow-up from the start, during, and after cancer treat-
ment is advised. An absolute drop of >10% in LVEF to <50% indicates cardiac
dysfunction. Global longitudinal strain is more sensitive to detect cardiac dys-
function at earlier stages. Monitoring of biomarkers such as (pro-NT)BNPs and
troponin is also advised, especially in patients on immunotherapy with immune
checkpoint inhibitors being at increased risk of myocarditis [100]. People who
survived cancer should remain monitored in the long term as HF may develop
several years after cancer treatment.
19 Heart Failure 259
19.6.3.3 Infections
Infections, viral or bacterial, may trigger acute heart failure and may worsen chronic
HF. Due to diminished activity of the immune system and underlying chronic disor-
ders such as COPD, older persons are more susceptible for infections. On top of
that, in the older patient, infection may lead to severe physical and mental dysregu-
lation and rapid global deterioration. Sepsis can cause myocardial damage and
depression leading to severe heart failure, with higher risk in people with pre-
existing heart failure.
Older people and people with cardiovascular morbidity have a higher risk for all
types of pneumonia (community acquired, healthcare related, and aspiration pneu-
monia) with increased risk for a severe course leading to respiratory and cardiac
failure. An observational retrospective study identified clinical characteristics and
risk factors of pulmonary infection in older patients with heart failure and found that
age ≥70 years, diabetes, NYHA III, LVEF <55%, and CRP ≥10 mg/L are higher
risks of pulmonary infections. Preventive measures targeting these risk factors
might reduce pulmonary infections [106]. Influenza and coronavirus disease 2019
(COVID-19 (SARS-COV-2)) are major causes of morbidity and mortality, includ-
ing heart failure. Course of the disease and case fatality rates are highest at older
ages [107–109]. The multicentre, multinational PCHF-COVICAV registry assessed
the outcome of hospitalised COVID-19 patients with heart failure (HF) compared
with patients with other cardiovascular disease and/or risk factors (arterial hyperten-
sion, diabetes, or dyslipidaemia). Hospitalised COVID-19 patients with HF were at
1.5-fold increased risk for in-hospital death. In-hospital worsening of HF or acute
HF de novo was common and associated with a further increase in in-hospital
mortality(threefold increased likelihood) [110]. In observational studies and retro-
spective analyses, influenza vaccination is associated with a reduced risk of all-
cause death in patients with HF [111]. Influenza, COVID-19 vaccination, and
pneumococcal vaccination should be considered in patients with HF.
19.7 Considerations
An increase in resource utilisation and economic costs are expected in the grow-
ing older population with multiple comorbidities and with broad discrepancies in
outcomes. Implementation of prospective databases, quality checks, and cost-
efficiency of treatment plans is essential [112].
The ESC 2021 cardiovascular disease prevention guidelines are a major step
forward by considering age more appropriately in the risk estimation scores
(SCORE2-OP) and in risk factor management (lifetime benefit estimation of treat-
ment), taking into account comorbidities and patient preferences.
Future guidelines should focus more on the ageing population, an ever-growing
yet highly heterogeneous population which makes a tailored holistic personal
approach more than necessary.
References
1. Unlu O, Levitan EB, Reshetnyak E, Kneifati-Hayek J, Diaz I, Archambault A,
et al. Polypharmacy in older adults hospitalized for heart failure. Circ Hear Fail.
2020;13(11):e006977.
2. Cherubini A, Laroche ML, Petrovic M. Mastering the complexity: drug therapy optimization
in geriatric patients. Eur Geriatr Med [Internet]. 2021;12(3):431–4. https://doi.org/10.1007/
s41999-021-00493-5.
3. McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, et al. 2021
ESC guidelines for the diagnosis and treatment of acute and chronic heart failure.
Eur Heart J [Internet]. 2021;42(36):3599–726. https://academic.oup.com/eurheartj/
article/42/36/3599/6358045.
4. Jankowska EA, Vitale C, Uchmanowicz I, Tkaczyszyn M, Drozd M, Ponikowski P. Drug
therapy in elderly heart failure patients. Eur Hear J Suppl. 2019;21:L8–L11.
5. Damy T, Chouihed T, Delarche N, Berrut G, Cacoub P, Henry P, et al. Diagnosis and man-
agement of heart failure in elderly patients from hospital admission to discharge: Position
paper. 2021.
6. Ceia F, Fonseca C, Mota T, Morais H, Matias F, De Sousa A, et al. Prevalence of chronic heart
failure in Southwestern Europe: the EPICA study. Eur J Heart Fail. 2002;4(4):531.
7. Van Riet EES, Hoes AW, Limburg A, Landman MAJ, Van Der Hoeven H, Rutten
FH. Prevalence of unrecognized heart failure in older persons with shortness of breath on
exertion. Eur J Heart Fail. 2014;16(7):772.
8. Bleumink GS, Knetsch AM, Sturkenboom MCJM, Straus SMJM, Hofman A, Deckers JW,
et al. Quantifying the heart failure epidemic: prevalence, incidence rate, lifetime risk and
prognosis of heart failure—the Rotterdam study. Eur Heart J. 2004;25(18):1614.
9. Koh AS, Tay WT, Teng THK, Vedin O, Benson L, Dahlstrom U, et al. A comprehensive
population-based characterization of heart failure with mid-range ejection fraction. Eur J
Heart Fail. 2017;19(12):1624.
10. Chioncel O, Lainscak M, Seferovic PM, Anker SD, Crespo-Leiro MG, Harjola VP, et al.
Epidemiology and one-year outcomes in patients with chronic heart failure and preserved,
mid-range and reduced ejection fraction: an analysis of the ESC heart failure long-term reg-
istry. Eur J Heart Fail. 2017;19(12):1574.
11. Heiat A, Gross CP, Krumholz HM. Representation of the elderly, women, and minorities in
heart failure clinical trials. Arch Intern Med. 2002;162:1682–8.
12. Gerber Y, Weston SA, Redfield MM, Chamberlain AM, Manemann SM, Jiang R, et al. A
contemporary appraisal of the heart failure epidemic in Olmsted County, Minnesota, 2000 to
2010. JAMA Intern Med. 2015;175(6):996–1004.
262 T. L. De Backer and A. A. Mangoni
13. Tsao CW, Lyass A, Enserro D, Larson MG, Ho JE, Kizer JR, et al. Temporal trends in the
incidence of and mortality associated with heart failure with preserved and reduced ejection
fraction. JACC Heart Fail. 2018;6(8):678.
14. Owan TE, Hodge DO, Herges RM, Jacobsen SJ, Roger VL, Redfield MM. Trends in
prevalence and outcome of heart failure with preserved ejection fraction. N Engl J Med.
2006;355(3):251.
15. Groenewegen A, Rutten FH, Mosterd A, Hoes AW. Epidemiology of heart failure. Eur J Heart
Fail. 2020;22:1342–56.
16. Shah KS, Xu H, Matsouaka RA, Bhatt DL, Heidenreich PA, Hernandez AF, et al. Heart
failure with preserved, borderline, and reduced ejection fraction: 5-year outcomes. J Am Coll
Cardiol. 2017;70(20):2476.
17. Buddeke J, Valstar GB, Van Dis I, Visseren FLJ, Rutten FH, Den Ruijter HM, et al. Mortality
after hospital admission for heart failure: improvement over time, equally strong in women
as in men. BMC Public Health. 2020;20(1):36.
18. Blais C, Dai S, Waters C, Robitaille C, Smith M, Svenson LW, et al. Assessing the burden
of hospitalized and community-care heart failure in Canada. Can J Cardiol. 2014;30(3):352.
19. Dai S, Walsh P, Wielgosz A, Gurevich Y, Bancej C, Morrison H. Comorbidities and mortality
associated with hospitalized heart failure in Canada. Can J Cardiol. 2012;28(1):74.
20. Guerra F, Brambatti M, Matassini MV, Capucci A. Current therapeutic options for heart fail-
ure in elderly patients. Biomed Res Int. 2017;2017:1483873. (Table 1).
21. Hill E, Taylor J. Chronic heart failure care planning: considerations in older patients. Card
Fail Rev. 2017;3(1):46.
22. Journal TP, Beezer BJ, Beezer J. Heart failure, older people and frailty diagnosis of heart
failure and frailty can often be masked by other long-term conditions, making the delivery.
2021;(July 2019):1–16.
23. Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, et al. Empagliflozin in
heart failure with a preserved ejection fraction. N Engl J Med. 2021;385(16):1451–61.
24. Mordi IR, Ouwerkerk W, Anker SD, Cleland JG, Dickstein K, Metra M, et al. Heart failure
treatment up-titration and outcome and age: an analysis of BIOSTAT-CHF. Eur J Heart Fail.
2021;23(3):436.
25. Japp D, Shah A, Fisken S, Denvir M, Shenkin S, Japp A. Mineralocorticoid receptor antag-
onists in elderly patients with heart failure: a systematic review and meta-analysis. Age
Ageing. 2017;46:18–25.
26. Ferreira JP, Rossello X, Eschalier R, McMurray JJV, Pocock S, Girerd N, et al. MRAs in
elderly HF patients: individual patient-data meta-analysis of RALES, EMPAHSIS-HF, and
TOPCAT. JACC Heart Fail. 2019;7(12):1012–21.
27. Desai RJ, Patorno E, Vaduganathan M, Mahesri M, Chin K, Levin R, et al. Effectiveness of
angiotensin-neprilysin inhibitor treatment versus renin-angiotensin system blockade in older
adults with heart failure in clinical care. Heart. 2021;107(17):1407.
28. Jhund PS, Fu M, Bayram E, Chen CH, Negrusz-Kawecka M, Rosenthal A, et al. Efficacy and
safety of LCZ696 (sacubitril-valsartan) according to age: insights from PARADIGM-HF. Eur
Heart J. 2015;36(38):2576.
29. Zannad F, Ferreira JP, Pocock SJ, Anker SD, Butler J, Filippatos G, et al. SGLT2 inhibitors in
patients with heart failure with reduced ejection fraction: a meta-analysis of the EMPEROR-
reduced and DAPA-HF trials. Lancet. 2020;396(10254):819.
30. Fioretto P, Bruin T, Johnsson E, Ptaszynska A, Parikh S, List JF. Safety and efficacy of the
SGLT2 inhibitor dapagliflozin in older patients with type 2 diabetes. Diabetologia. 2013;56.
31. Cleland JGF, Tendera M, Adamus J, Freemantle N, Polonski L, Taylor J. The per-
indopril in elderly people with chronic heart failure (PEP-CHF) study. Eur Heart
J. 2006;27(19):2338–45.
32. Flather MD, Shibata MC, Coats AJS, Van Veldhuisen DJ, Parkhomenko A, Borbola J, et al.
FASTTRACK randomized trial to determine the effect of nebivolol on mortality and car-
diovascular hospital admission in elderly patients with heart failure (SENIORS). Eur Heart
J. 2005;26(3):215.
19 Heart Failure 263
33. Düngen HD, Apostolović S, Inkrot S, Tahirović E, Töpper A, Mehrhof F, et al. Titration to
target dose of bisoprolol vs. carvedilol in elderly patients with heart failure: the CIBIS-ELD
trial. Eur J Heart Fail. 2011;13(6):670.
34. Düngen HD, Musial-Bright L, Inkrot S, Apostolovic̈ S, Edelmann F, Lainščak M, et al. Heart
rate following short-term beta-blocker titration predicts all-cause mortality in elderly chronic
heart failure patients: insights from the CIBIS-ELD trial. Eur J Heart Fail. 2014;16(8):907.
35. Yokote K, Terauchi Y, Nakamura I, Sugamori H. Real-world evidence for the safety of ipra-
gliflozin in elderly Japanese patients with type 2 diabetes mellitus (STELLA-ELDER): final
results of a post-marketing surveillance study. Expert Opin Pharmacother. 2016;17(15):1995.
36. Kambara T, Shibata R, Osanai H, Nakashima Y, Asano H, Sakai K, et al. Use of sodium–
glucose cotransporter 2 inhibitors in older patients with type 2 diabetes mellitus. Geriatr
Gerontol Int. 2018;18(1):108.
37. Trescoli Serrano C, Fajardo CJ, Arazo J, Tarazona-Santabalbina FJ. Real world safety
and efficacy of SGLT-2 inhibitors in elderly type 2 diabetic patients. Innov Aging.
2017;1(Suppl 1):148.
38. Yabe D, Shiki K, Suzaki K, Meinicke T, Kotobuki Y, Nishida K, et al. Rationale and design
of the EMPA-ELDERLY trial: a randomised, double-blind, placebo-controlled, 52-week
clinical trial of the efficacy and safety of the sodium-glucose cotransporter-2 inhibitor empa-
gliflozin in elderly Japanese patients with type 2 diabetes. BMJ Open. 2021;11(4):e045844.
39. Karagiannis T, Tsapas A, Athanasiadou E, Avgerinos I, Liakos A, Matthews DR, et al. GLP-1
receptor agonists and SGLT2 inhibitors for older people with type 2 diabetes: a systematic
review and meta-analysis. Diabetes Res Clin Pract. 2021;174:108737.
40. Armstrong PW, Roessig L, Patel MJ, Anstrom KJ, Butler J, Voors AA, et al. A multicenter,
randomized, double-blind, placebo-controlled trial of the efficacy and safety of the oral sol-
uble guanylate cyclase stimulator: the VICTORIA trial. JACC Heart Fail. 2018;6(2):96–104.
41. Screever EM, Meijers WC, van Veldhuisen DJ, de Boer RA. New developments in the phar-
macotherapeutic management of heart failure in elderly patients: concerns and consider-
ations. Expert Opin Pharmacother. 2017;18:645.
42. Patel PH, Nguyen M, Rodriguez R, Surani S, Udeani G. Omecamtiv mecarbil: a novel
mechanistic and therapeutic approach to chronic heart failure management. Cureus.
2021;13(1):e12419.
43. Teerlink JR, Diaz R, Felker GM, McMurray JJV, Metra M, Solomon SD, et al. Omecamtiv
mecarbil in chronic heart failure with reduced ejection fraction: rationale and design of
GALACTIC-HF. JACC Heart Fail. 2020;8:329.
44. Swedberg K. Stimulation of contractility in systolic heart failure. N Engl J Med.
2021;384(2):178.
45. Visseren FLJ, Mach F, Smulders YM, Carballo D, Koskinas KC, Bäck M, et al. 2021 ESC
guidelines on cardiovascular disease prevention in clinical practice. Eur Heart J [Internet].
2021;42(34):3227–3337. https://academic.oup.com/eurheartj/article/42/34/3227/6358713.
46. Burnier M, Egan BM. Adherence in hypertension: a review of prevalence, risk factors,
impact, and management. Circ Res. 2019;124(7):1124.
47. OH, JFA, AB, GB, JPE, PF, et al. Guidelines for the management and diagnosis of congestive
heart failure in the elderly adult. Rev Geriatr. 2004;29(8).
48. Abraham WT, Fonarow GC, Albert NM, Stough WG, Gheorghiade M, Greenberg BH, et al.
Predictors of in-hospital mortality in patients hospitalized for heart failure. Insights from
the Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients with Heart
Failure (OPTIMIZE-HF). J Am Coll Cardiol. 2008;52(5):347–56.
49. Detremerie C, De Backer T, Petrovic M. Polypharmacy and management of hypertension in
frail elderly. Tijdschr Geneeskd. 2017;73(3).
50. Benetos A, Rossignol P, Cherubini A, Joly L, Grodzicki T, Rajkumar C, et al. Polypharmacy
in the aging patient: management of hypertension in octogenarians. JAMA J Am Med Assoc.
2015;314(2):170.
51. Williams B, Mancia G, Spiering W, Rosei EA, Azizi M, Burnier M, et al. 2018 ESC/ESH
guidelines for the management of arterial hypertension. J Hypertens. 2018;36:1956–2041.
264 T. L. De Backer and A. A. Mangoni
70. Sartipy U, Savarese G, Dahlström U, Fu M, Lund LH. Association of heart rate with mortal-
ity in sinus rhythm and atrial fibrillation in heart failure with preserved ejection fraction. Eur
J Heart Fail. 2019;21(4):471.
71. Cho SW, Bin GH, Hwang JK, Chun KJ, Park KM, On YK, et al. Clinical features, pre-
dictors, and long-term prognosis of pacing-induced cardiomyopathy. Eur J Heart Fail.
2019;21(5):643.
72. St. John Sutton M, Plappert T, Adamson PB, Li P, Christman SA, Chung ES, et al. Left
ventricular reverse remodeling with biventricular versus right ventricular pacing in patients
with atrioventricular block and heart failure in the BLOCK HF trial. Circ Heart Fail.
2015;8(3):510–8.
73. Abdelrahman M, Subzposh FA, Beer D, Durr B, Naperkowski A, Sun H, et al. Clinical
outcomes of his bundle pacing compared to right ventricular pacing. J Am Coll Cardiol.
2018;71(20):2319.
74. Ravi V, Beer D, Pietrasik GM, Hanifin JL, Ooms S, Ayub MT, et al. Development of
new-onset or progressive atrial fibrillation in patients with permanent his bundle pacing
versus right ventricular pacing: results from the RUSH HBP registry. J Am Heart Assoc.
2020;9(22):e018478.
75. Fauchier L, Alonso C, Anselme F, Blangy H, Bordachar P, Boveda S, et al. Position paper
for management of elderly patients with pacemakers and implantable cardiac defibrilla-
tors Groupe de rythmologie et stimulation cardiaque de la Société française de cardiolo-
gie et Société française de gériatrie et gérontologie. Gériatrie Psychol Neuropsychiatr du
Viellissement. 2019;14(3):239.
76. Vaes B, Rezzoug N, Pasquet A, Wallemacq P, Van Pottelbergh G, Matheï C, et al. The prev-
alence of cardiac dysfunction and the correlation with poor functioning among the very
elderly. Int J Cardiol. 2012;155(1):134.
77. Stewart BF, Siscovick D, Lind BK, Gardin JM, Gottdiener JS, Smith VE, et al. Clinical fac-
tors associated with calcific aortic valve disease. J Am Coll Cardiol. 1997;29(3):630.
78. Osnabrugge RLJ, Mylotte D, Head SJ, Van Mieghem NM, Nkomo VT, Lereun CM, et al.
Aortic stenosis in the elderly: disease prevalence and number of candidates for transcath-
eter aortic valve replacement: a meta-analysis and modeling study. J Am Coll Cardiol.
2013;62(11):1002.
79. Généreux P, Stone GW, O’Gara PT, Marquis-Gravel G, Redfors B, Giustino G, et al. Natural
history, diagnostic approaches, and therapeutic strategies for patients with asymptomatic
severe aortic stenosis. J Am Coll Cardiol. 2016;67:2263.
80. Vahanian A, Beyersdorf F, Praz F, Milojevic M, Baldus S, Bauersachs J, et al. 2021
ESC/EACTS guidelines for the management of valvular heart disease. Eur Heart
J. 2022;43(7):561–632.
81. Afilalo J, Mottillo S, Eisenberg MJ, Alexander KP, Noiseux N, Perrault LP, et al. Addition of
frailty and disability to cardiac surgery risk scores identifies elderly patients at high risk of
mortality or major morbidity. Circ Cardiovasc Qual Outcomes. 2012;5(2):222–8.
82. Leon MB, Smith CR, Mack M, Miller DC, Moses JW, Svensson LG, et al. Transcatheter
aortic-valve implantation for aortic stenosis in patients who cannot undergo surgery. N Engl
J Med. 2010;363(17):1597.
83. Smith CR, Leon MB, Mack MJ, Miller DC, Moses JW, Svensson LG, et al. Transcatheter ver-
sus surgical aortic-valve replacement in high-risk patients. Surv Anesthesiol. 2012;56(1):4.
84. Makkar RR, Fontana GP, Jilaihawi H, Kapadia S, Pichard AD, Douglas PS, et al.
Transcatheter aortic-valve replacement for inoperable severe aortic stenosis. N Engl J Med.
2012;366(18):1696.
85. Bertrand PB, Schwammenthal E, Levine RA, Vandervoort PM. Exercise dynamics in sec-
ondary mitral regurgitation: pathophysiology and therapeutic implications. Circulation.
2017;135:297.
86. Dziadzko V, Clavel MA, Dziadzko M, Medina-Inojosa JR, Michelena H, Maalouf J, et al.
Outcome and undertreatment of mitral regurgitation: a community cohort study. Lancet.
2018;391(10124):960.
266 T. L. De Backer and A. A. Mangoni
87. Goliasch G, Bartko PE, Pavo N, Neuhold S, Wurm R, Mascherbauer J, et al. Refining the
prognostic impact of functional mitral regurgitation in chronic heart failure. Eur Heart
J. 2018;39(1):39.
88. Ghoreishi M, Dawood MY, Gammie JS. Mitral valve surgery in elderly patients with mitral
regurgitation: repair or replacement with tissue valve? Curr Opin Cardiol. 2013;28:164.
89. Farid S, Ladwiniec A, Hernández-Sánchez J, Povey H, Caruana E, Ali A, et al. Early out-
comes after mitral valve repair versus replacement in the elderly: a propensity matched analy-
sis. Heart Lung Circ. 2019;28(2):314–9.
90. Feldman T, Foster E, Glower DD, Kar S, Rinaldi MJ, Fail PS, et al. Percutaneous repair or
surgery for mitral regurgitation. N Engl J Med. 2011;364(15):1395.
91. Coats AJS, Anker SD, Baumbach A, Alfieri O, von Bardeleben RS, Bauersachs J, et al. The
management of secondary mitral regurgitation in patients with heart failure: a joint position
statement from the Heart Failure Association (HFA), European Association of Cardiovascular
Imaging (EACVI), European Heart Rhythm Association (EHRA), and European
Association of Percutaneous Cardiovascular Interventions (EAPCI) of the ESC. Eur Heart
J. 2021;42(13):1254–69.
92. Ruf TF, Kreidel F, Tamm AR, Geyer M, Hahad O, Zirbs JC, et al. Transcatheter indirect
mitral annuloplasty induces annular and left atrial remodelling in secondary mitral regurgita-
tion. ESC Heart Fail. 2020;7(4):1400.
93. Geyer M, Keller K, Sotiriou E, Tamm AR, Ruf TF, Kreidel F, et al. Association of transcath-
eter direct mitral annuloplasty with acute anatomic, haemodynamic, and clinical outcomes in
severe mitral valve regurgitation. ESC Heart Fail. 2020;7(6):3336.
94. Giallauria F, Di Lorenzo A, Parlato A, Testa C, Bobbio E, Vigorito C, et al. Individual patient
data meta-analysis of the effects of the CARILLON® mitral contour system. ESC Heart Fail.
2020;7(6):3383.
95. Malik AH, Zaid S, Yandrapalli S, Shetty S, Aronow WS, Ahmad H, et al. Trends and out-
comes with Transcatheter versus surgical mitral valve repair in patients ≥80 years of age. Am
J Cardiol. 2020;125(7):1083.
96. Del Val D, Ferreira-Neto AN, Wintzer-Wehekind J, Dagenais F, Paradis JM, Bernier M, et al.
Early experience with transcatheter mitral valve replacement: a systematic review. J Am
Heart Assoc. 2019;8(17):e013332.
97. Fiorilli PN, Herrmann HC, Szeto WY. Transcatheter mitral valve replacement: latest advances
and future directions. Ann Cardiothorac Surg. 2021;10(1):85.
98. Reddy P, Shenoy C, Blaes AH. Cardio-oncology in the older adult. J Geriatr Oncol.
2017;8:308.
99. Lyon AR, Dent S, Stanway S, Earl H, Brezden-Masley C, Cohen-Solal A, et al. Baseline car-
diovascular risk assessment in cancer patients scheduled to receive cardiotoxic cancer thera-
pies: a position statement and new risk assessment tools from the Cardio-Oncology Study
Group of the Heart Failure Association of the European Society of Cardiology in collabora-
tion with the International Cardio-Oncology Society. Eur J Heart Fail. 2020;22(11):1945–60.
100. Anker MS, Von Haehling S, Anker SD. Novel biomarkers in heart failure and cardio-
oncology. Kardiol Pol. 2019;77:329.
101. McDonagh T, Damy T, Doehner W, Lam CSP, Sindone A, van der Meer P, et al. Screening,
diagnosis and treatment of iron deficiency in chronic heart failure: putting the 2016 European
Society of Cardiology heart failure guidelines into clinical practice. Eur J Heart Fail.
2018;20:1664.
102. Mini P, Marc B, Fabien S, Chuzeville M. Prevalence of iron deficiency in patients aged 75
years or older with heart failure. J Geriatr Cardiol. 2018;15(11):682–6.
103. von Haehling S, Ebner N, Evertz R, Ponikowski P, Anker SD. Iron deficiency in heart failure:
an overview. JACC Heart Fail. 2019;7:36–46.
104. Stugiewicz M, Tkaczyszyn M, Kasztura M, Banasiak W, Ponikowski P, Jankowska EA. The
influence of iron deficiency on the functioning of skeletal muscles: experimental evidence
and clinical implications. Eur J Heart Fail. 2016;18:762.
19 Heart Failure 267
105. Ponikowski P, Macdougall IC, Bohm M, Comin Colet J, Filippatos G, Roubert B, et al. An
individual patient data meta-analysis of outcomes in 4 randomized double-blind trials of iron-
deficient patients with HFrEF and renal dysfunction treated with IV ferric carboxymaltose.
Eur J Heart Fail. 2017;19.
106. Peng Q, Yang Q. Risk factors and management of pulmonary infection in elderly patients
with heart failure. Medicine (Baltimore). 2021;100(38):e27238.
107. Tomasoni D, Italia L, Adamo M, Inciardi RM, Lombardi CM, Solomon SD, et al. COVID-19
and heart failure: from infection to inflammation and angiotensin II stimulation. Searching
for evidence from a new disease. Eur J Heart Fail. 2020;22:957.
108. Rey JR, Caro-Codón J, Rosillo SO, Iniesta AM, Castrejón-Castrejón S, Marco-Clement I,
et al. Heart failure in COVID-19 patients: prevalence, incidence and prognostic implications.
Eur J Heart Fail. 2020;22(12):2205.
109. Vardeny O, Solomon SD. Influenza and heart failure: a catchy comorbid combination. JACC
Heart Fail. 2019;7:118–20.
110. Sokolski M, Trenson S, Sokolska JM, D’Amario D, Meyer P, Poku NK, et al. Heart failure
in COVID-19: the multicentre, multinational PCHF-COVICAV registry, vol. 8. ESC Heart
Fail; 2021. p. 4955.
111. Modin D, Jørgensen ME, Gislason G, Jensen JS, Køber L, Claggett B, et al. Influenza vaccine
in heart failure. Circulation. 2019;139(5):575–86.
112. Iyngkaran P, Liew D, Neil C, Driscoll A, Marwick TH, Hare DL. Moving from heart failure
guidelines to clinical practice: gaps contributing to readmissions in patients with multiple
comorbidities and older age. Clin Med Insights Cardiol. 2018;12:1179546818809358.
Pharmacological Treatment of Cognitive
and Behavioral Disorders in Dementia 20
F. Trotta, L. Biscetti, and A. Cherubini
20.1 Introduction
The most common (60–80% of all dementia cases) is Alzheimer’s disease (AD).
AD has a complex etiology, and our understanding of the relationships between the
numerous pathways involved in the development of AD and subsequent neurode-
generation is still incomplete. AD is characterized by extracellular amyloid plaques,
intracellular neurofibrillary tangles, and nerve cell death [4–6]. The typical form of
Alzheimer’s disease is characterized by a progressive episodic memory loss, subse-
quently accompanied by cognitive deficits involving non-amnestic domains, e.g.,
language and executive functions.
Vascular dementia (VaD) is considered the second most common cause of dementia
in the elderly. VaD is defined as any form of dementia caused by cerebrovascular
disease or impaired blood flow. VaD can develop after stroke and can have a step-
wise progression; otherwise, it can develop in individuals without clinical strokes
and can have a gradual progression [7].
and 10.2% in those aged <65 [10–12]. There are different types of frontotemporal
dementia, depending on which part of the brain is affected: behavioral variant (fron-
tal) and primary progressive aphasia.
Currently, in the management of dementia, the main goals are to treat symptoms
related to cognitive decline and to reduce the burden of the disease on caregivers.
The recommended treatment strategy includes symptomatic pharmacological ther-
apy of cognitive deficits, cognitive rehabilitation, environmental adaptations to
reduce the impact of cognitive deficits and behavioral symptoms, nonpharmacologi-
cal and pharmacological treatment of behavioral symptoms, and prevention and
treatment of complications (e.g., falls, malnutrition, incontinence, bed rest; plan-
ning of care and family support; education and support of caregivers).
First of all, it is necessary to evaluate the medical history: The presence of
drugs with anticholinergic effect can induce a worsening of cognitive functions.
An increased risk for dementia was seen in people with higher use of anticho-
linergics (e.g., antihistamines, gastrointestinal tract antispasmodics, antivertigo
agents/antiemetic, antidepressants, bladder antimuscarinics, skeletal muscle
relaxant, antipsychotics, antiarrhythmic, antiparkinsonians). Therefore, pre-
scribers should be aware of this potential association, and alternative drugs
should be considered when possible. If no therapeutic alternative exists, pre-
scribers should use the lowest effective dose and discontinue therapy if ineffec-
tive [13]. Some tools have been developed to quantify the cumulative
anticholinergic effect of the drugs and their impact on cognitive functions and
activities of life: Anticholinergic Drug Scale (ADS), the Anticholinergic Burden
Scale (ACS), the Anticholinergic Risk Scale (ARS), and the Drug Burden Index
(DBI) [14–17]. In the advanced stages of dementia, the worsening of the impair-
ment of cognitive functions makes it impossible to carry out the activities of
daily living. This leads to an increased risk of malnutrition, pneumonia, and
pressure sores. As the disease progresses, treatment should be aimed to preserve
the patient’s well-being, rather than trying to prolong life. Provision of pallia-
tive care was empirically recognized as a care step in the management of demen-
tia. However, as several studies have shown, the palliative care pathway for
patients with dementia is still far from being accepted, shared, and applied [18–
20]. Holmes criteria have been developed for the appropriateness of prescribing
drugs in advanced dementia [21].
272 F. Trotta et al.
20.2.2 Memantine
20.2.3 Aducanumab
• Mild cognitive impairment or mild AD (MMSE ≥21, MoCA ≥17, or CDR 0.5–1)
• Documented amyloid pathology: Clinicians should limit use of aducanumab to
those patients proven to be amyloid positive (by amyloid positron emission
tomography [PET] scan or lumbar puncture)
Other agents have been studied for VaD, but the trials have mostly been small and
without conclusive results [65]: nimodipine, ergot alkaloids (hydergine, nicergo-
line), cerebrolysin, vitamin E, ginkgo biloba, xanthine derivatives (propentofylline,
pentoxifylline, and denbufylline), cytidinediphosphocholine, and piracetam. None
of these therapies are recommended at this time.
Apathy, depression, anxiety, and agitation were found to be the most frequent
forms of BPSD. However, a recent systematic review revealed substantial variation
in the reported prevalence, incidence, and longitudinal course among different stud-
ies. In an individual patient, the type and severity of BPSD tend to change over time,
but some forms such as wandering seem to be more persistent. Overall, the “natural
course” of BPSD over time is still largely unknown [70].
The treatment of BPSD is often highly challenging. BPSD management requires
both a patient-centered and caregiver-centered focus. Treating concomitant somatic
diseases can reduce BPSD. Effective pain management is part of successful BPSD
treatment. It is important to always consider medication toxicity, such as anticholin-
ergic side effects, or abrupt discontinuation of chronic therapies. Pharmacotherapy
for BPSD is frequently provided, but it carries the risk of serious side effects.
Therefore, nonpharmacological therapies are the first choice, and they should be
continued also when pharmacotherapy is necessary.
20 Pharmacological Treatment of Cognitive and Behavioral Disorders in Dementia 279
The hyperactivity-impulsivity-irritability-disinhibition-aggression-agitation
(HIDA) domain represents one of the most difficult sets of symptoms to manage
and accounts for much of the burden for caregivers and hospital staff. These symp-
toms can arise from a variety of underlying causes in patients with dementia, and
identifying the genesis of the abnormal behavior is critical to effective management
(e.g., delirium, depression, sleep disorders, misperception). Many studies suggest
non-pharmacological treatments for HIDA domain symptoms as first line, for
instance, music therapy, sensory interventions, and person-centered communication
skill for caregivers [71].
When non-pharmacological intervention fails and neuropsychiatric symptoms
produced severe distress or safety issues, acute pharmacologic therapy with an anti-
psychotic drug is often necessary [72]. NICE Guidelines recommend (1) to use the
lowest effective dose for the shortest possible time, (2) to reassess the patients at
least every 6 weeks, (3) to check whether they still need medication, and (4) to stop
treatment with antipsychotics if the person with dementia is not getting a clear
ongoing benefit from taking these drugs (after discussion with the person taking
them and their family members or careers when appropriate). To this regard, it
should be acknowledged that many of these drugs may increase mortality, and most
of them are not approved for treatment of behavioral disorders in patients with
dementia by FDA. Despite the warnings issued by the FDA, the EMA and the UK
Medicines and Healthcare Products Regulatory Agency mainly related to increased
risk of death, cerebrovascular adverse events (CVAEs), parkinsonism, sedation, gait
disturbance, cognitive decline, and pneumonia, antipsychotics are often used in
individuals with dementia for sustained periods (≥6 months). It is noteworthy that
the drug-related mortality risk remains elevated for at least 2 years, with a risk of
death that is greater than the longer duration of use [69].
Benzodiazepines are not recommended for the management of neuropsychiatric
symptoms in elderly patients with dementia because they can induce worsening of
gait and balance, potential paradoxical agitation, and physical dependence [73].
Risperidone
Risperidone, at low dosages (1–2 mg per day), shows higher affinity for 5-HT 2A
than for D2, while at high dosages (3–6 mg per day), the affinity for D2 overcomes
that for 5-HT2A. Risperidone has shown good efficacy in treating positive symp-
toms. However, the strong binding to 5-HT 2C, α1, and H1 is responsible for the
side effects, such as weight gain, sedation, orthostatic hypotension, and
parkinsonism.
Aripiprazole
Together with amisulpride, aripiprazole represents an exception among AAPs,
given the relatively low affinity to 5HT2A receptor. It is associated to a low risk of
extrapyramidal side effects, and it is efficacious against both positive and negative
symptoms. Aripiprazole causes minimal weight gain and sedation and does not pro-
duce elevation in serum prolactin levels; most importantly, unlike other neurolep-
tics, it does not significantly lengthen QTc interval. However, based on randomized
controlled trial performed to test efficacy and safety of aripiprazole in the context of
Alzheimer’s dementia [77], faced to a small but statistically significant efficacy in
controlling BPSD in AD patients, aripiprazole resulted to be associated to higher
mortality and higher risk of stroke compared to placebo. Although a real causal role
of the drug in inducing these severe side effects is still controversial, the prevalent
expert opinion suggests that aripiprazole should be used with caution in AD
patients [78].
Olanzapine
Olanzapine can be started at a dose of 2.5 mg daily and titrated up to a maximum of
5 mg twice a day (the incidence of extrapyramidal symptoms is quite low at doses
of 5 mg per day). It appears to be quite effective for treating neuropsychiatric symp-
toms in Alzheimer’s disease or vascular dementia [79]. Metabolic side effects
(weight gain, diabetes, and hypercholesterolemia) are more common compared to
other antipsychotic drugs.
Quetiapine
Quetiapine generally is used at a dose of 25 mg at bedtime and titrating up a maxi-
mum of 75 mg twice a day. It has different effects based on the dose administrated:
antipsychotic effect with 600 mg per day, antidepressant with 300 mg per day, hyp-
notic effect with 50 mg per day. Because of its effects on the cardiac repolarization,
it is necessary to check the QTc before the prescription.
20 Pharmacological Treatment of Cognitive and Behavioral Disorders in Dementia 283
In conclusion, it must be highlighted that only three drugs are licensed for BPSD
treatment in dementia: pimavanserin (only in the USA) and clozapine for hallucina-
tions and delusions associated with Parkinson’s disease (PD) psychosis and risperi-
done for short-term treatment of persistent aggression in moderate-to-severe
Alzheimer’s disease (AD). In the remaining cases, the use of antipsychotics is off-
label and guided by the physician’s judgment.
Risperidone, aripiprazole, and olanzapine have been evaluated in multiple stud-
ies and show improvement in symptoms like severe agitation, aggression, and psy-
chosis (such as delusions and hallucinations) in patients with dementia.
Maximal duration of antipsychotic treatment should be limited according to 3Ts
(target symptom, titration, time) whereby the duration of the treatment should not
exceed 3 months due to an increased risk of mortality, and regular reassessment is
necessary.
Antipsychotics are generally to be avoided when diagnosing dementia with
Lewy bodies due to particular sensitivity of these patients to extrapyramidal side
effects. A use with caution of quetiapine and clozapine is possible [80, 81].
References
1. American Psychiatric Association. Diagnostic and statistical manual of mental disorders, fifth
edition (DSM-5). American Psychiatric Publishing; 2013.
2. Hugo J, Ganguli M. Dementia and cognitive impairment: epidemiology, diagnosis, and treat-
ment. Clin Geriatr Med. 2014;30(3):421–42.
3. Prince M, Wimo A, Guerchet M, Ali GC, Wu YT, Prina M. World Alzheimer report 2015, the
global impact of dementia: an analysis of prevalence, incidence, cost and trends. Alzheimer’s
Disease International; 2015.
4. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol
Med. 2016;8(6):595–608.
5. Iqbal K, del C Alonso A, Chen S, Chohan MO, El-Akkad E, Gong CX, Khatoon S, Li B, Liu
F, Rahman A, Tanimukai H, Grundke-Iqbal I. Tau pathology in Alzheimer disease and other
tauopathies. Biochim Biophys Acta. 2005;1739(2–3):198–210.
6. Mufson EJ, Counts SE, Perez SE, Ginsberg SD. Cholinergic system during the progression of
Alzheimer’s disease: therapeutic implications. Expert Rev Neurother. 2008;8(11):1703–18.
7. Wolters FJ, Ikram MA. Epidemiology of vascular dementia. Arterioscler Thromb Vasc Biol.
2019;39(8):1542–9.
8. Erkkinen MG, Kim MO, Geschwind MD. Clinical neurology and epidemiology of the major
neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2018;10(4):a033118.
9. The Lund and Manchester Groups. Clinical and neuropathological criteria for frontotemporal
dementia. J Neurol Neurosurg Psychiatry. 1994;57(4):416.
10. Lopez OL, Kuller LH. Epidemiology of aging and associated cognitive disorders: preva-
lence and incidence of Alzheimer’s disease and other dementias. Handb Clin Neurol.
2019;167:139–48.
11. Oh ES, Rabins PV. Dementia. Ann Intern Med. 2019;171(5):ITC33–48.
12. Arvanitakis Z, Shah RC, Bennett DA. Diagnosis and management of dementia: review.
JAMA. 2019;322(16):1589–99.
13. Gray SL, Anderson ML, Dublin S, Hanlon TJ, Hubbard R, Walker R, Yu O, Crane PK, Larson
EB. Cumulative use of strong anticholinergics and incident dementia: a prospective cohort
study. JAMA Intern Med. 2015;175(3):401–7.
284 F. Trotta et al.
14. Rudolph JL, Salow MJ, Angelini MC, McGlinchey RE. The anticholinergic risk scale and
anticholinergic adverse effects in older persons. Arch Intern Med. 2008;168(5):508–13.
15. O’Connell J, Burke É, Mulryan N, O’Dwyer C, Donegan C, McCallion P, McCarron M,
Henman MC, O’Dwyer M. Drug burden index to define the burden of medicines in older adults
with intellectual disabilities: an observational cross-sectional study. Br J Clin Pharmacol.
2018;84(3):553–67.
16. Salahudeen MS, Duffull SB, Nishtala PS. Anticholinergic burden quantified by anticholiner-
gic risk scales and adverse outcomes in older people: a systematic review. BMC Geriatrics.
2015;15:31.
17. Sargent L, Nalls M, Amella EJ, Mueller M, Lageman SK, Bandinelli S, Colpo M, Slattum PW,
Singleton A, Ferrucci L. Anticholinergic drug induced cognitive and physical impairment:
results from the InCHIANTI Study. J Gerontol A Biol Sci Med Sci. 2020;75(5):995–1002.
18. Mataqi M, Aslanpour Z. Factors influencing palliative care in advanced dementia: a systematic
review. BMJ Support Palliat Care. 2020;10(2):145–56.
19. Mitchell SL, Teno JM, Kiely DK, Shaffer ML, Jones RN, Prigerson HG, Volicer L, Givens JL,
Hamel MB. The clinical course of advanced dementia. N Engl J Med. 2009;361(16):1529–38.
20. Sampson EL, Candy B, Davis S, Gola AB, Harrington J, King M, Kupeli N, Leavey G, Moore
K, Nazareth I, Omar RZ, Vickerstaff V, Jones L. Living and dying with advanced dementia:
a prospective cohort study of symptoms, service use and care at the end of life. Palliat Med.
2018;32(3):668–81.
21. Holmes HM, Sachs GA, Shega JW, Hougham GW, Cox HD, Dale W. Integrating palliative
medicine into the care of persons with advanced dementia: identifying appropriate medication
use. J Am Geriatr Soc. 2008;56:1306–11.
22. Abraha I, Rimland JM, Trotta FM, Dell’Aquila G, Cruz-Jentoft A, Petrovic M, Gudmundsson
A, Soiza R, O’Mahony D, Guaita A, Cherubini A. Systematic review of systematic reviews
of non-pharmacological interventions to treat behavioural disturbances in older patients with
dementia. The SENATOR-OnTop series. BMJ Open. 2017;7(3):e012759.
23. Wilcock G, Howe I, Coles H, Lilienfeld S, Truyen L, Zhu Y, Bullock R, Kershaw P, GAL-
GBR-2 Study Group. A long-term comparison of galantamine and donepezil in the treatment
of Alzheimer’s disease. Drugs Aging. 2003;20(10):777.
24. Ancoli-Israel S, Amatniek J, Ascher S, Sadik K, Ramaswamy K. Effects of galantamine ver-
sus donepezil on sleep in patients with mild to moderate Alzheimer disease and their care-
givers: a double-blind, head-to-head, randomized pilot study. Alzheimer Dis Assoc Disord.
2005;19(4):240.
25. Bullock R, Touchon J, Bergman H, Gambina G, He Y, Rapatz G, Nagel J, Lane R. Rivastigmine
and donepezil treatment in moderate to moderately-severe Alzheimer’s disease over a 2-year
period. Curr Med Res Opin. 2005;21(8):1317.
26. Campbell NL, Perkins AJ, Gao S, Skaar TC, Li L, Hendrie HC, Fowler N, Callahan CM,
Boustani MA. Adherence and tolerability of Alzheimer’s disease medications: a pragmatic
randomized trial. J Am Geriatr Soc. 2017;65(7):1497.
27. Chodosh J, Colaiaco BA, Connor KI, et al. Dementia care management in an underserved
community: the comparative effectiveness of two different approaches. J Aging Health.
2015;27(5):864–93.
28. Birks J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst Rev.
2006;2006(1):CD005593.
29. Xu H, Garcia-Ptacek S, Jönsson L, Wimo A, Nordström P, Eriksdotter M. Long-term effects of
cholinesterase inhibitors on cognitive decline and mortality. Neurology. 2021;96(17):e2220–30.
30. Zuin M, Cherubini A, Volpato S, Ferrucci L, Zuliani G. Acetyl-cholinesterase-inhibitors slow
cognitive decline and decrease overall mortality in older patients with dementia. Sci Rep.
2022;12(1):12214.
31. Cacabelos R. Pharmacogenetic considerations when prescribing cholinesterase inhibitors for
the treatment of Alzheimer’s disease. Expert Opin Drug Metab Toxicol. 2020;16(8):673–701.
32. Gorelick PB, Scuteri A, Black SE, Decarli C, Greenberg SM, Iadecola C, Launer LJ, Laurent S,
Lopez OL, Nyenhuis D, Petersen RC, Schneider JA, Tzourio C, Arnett DK, Bennett DA, Chui
20 Pharmacological Treatment of Cognitive and Behavioral Disorders in Dementia 285
HC, Higashida RT, Lindquist R, Nilsson PM, Roman GC, Sellke FW, Seshadri S, American
Heart Association Stroke Council, Council on Epidemiology and Prevention, Council on
Cardiovascular Nursing, Council on Cardiovascular Radiology and Intervention, and Council
on Cardiovascular Surgery and Anesthesia. Vascular contributions to cognitive impairment
and dementia: a statement for healthcare professionals from the American Heart Association/
American Stroke Association. Stroke. 2011;42(9):2672–713.
33. Kalaria RN, Ballard C. Overlap between pathology of Alzheimer disease and vascular demen-
tia. Alzheimer Dis Assoc Disord. 1999;13(Suppl 3):S115.
34. Meng YH, Wang PP, Song YX, Wang JH. Cholinesterase inhibitors and memantine for
Parkinson’s disease dementia and Lewy body dementia: a meta-analysis. Exp Ther Med.
2019;17(3):1611–24.
35. Mendez MF, Shapira JS, McMurtray A, Licht E. Preliminary findings: behavioral worsening on
donepezil in patients with frontotemporal dementia. Am J Geriatr Psychiatry. 2007;15(1):84.
36. Kimura T, Takamatsu J. Pilot study of pharmacological treatment for frontotemporal dementia:
risk of donepezil treatment for behavioral and psychological symptoms. Geriatr Gerontol Int.
2013;13(2):506.
37. Arciniegas DB, Anderson CA. Donepezil-induced confusional state in a patient with autopsy-
proven behavioral-variant frontotemporal dementia. J Neuropsychiatry Clin Neurosci.
2013;25(3):E25.
38. Kertesz A, Morlog D, Light M, Blair M, Davidson W, Jesso S, Brashear R. Galantamine in
frontotemporal dementia and primary progressive aphasia. Dement Geriatr Cogn Disord.
2008;25(2):178.
39. Moretti R, Torre P, Antonello RM, Cattaruzza T, Cazzato G, Bava A. Rivastigmine in fronto-
temporal dementia: an open-label study. Drugs Aging. 2004;21(14):931.
40. Hernandez RK, Farwell W, Cantor MD, Lawler EV. Cholinesterase inhibitors and incidence of
bradycardia in patients with dementia in the veterans affairs new England healthcare system. J
Am Geriatr Soc. 2009;57(11):1997.
41. Gill SS, Anderson GM, Fischer HD, Bell CM, Li P, Normand SL, Rochon PA. Syncope and
its consequences in patients with dementia receiving cholinesterase inhibitors: a population-
based cohort study. Arch Intern Med. 2009;169(9):867.
42. Birks JS, Harvey RJ. Donepezil for dementia due to Alzheimer’s disease (Review). Cochrane
Database Syst Rev. 2018;6(6):CD001190.
43. Loy C, Schneider L. Galantamine for Alzheimer’s disease and mild cognitive impairment.
Cochrane Database Syst Rev. 2006;2006(1):CD001747.
44. Winblad B, Cummings J, Andreasen N, Grossberg G, Onofrj M, Sadowsky C, Zechner S,
Nagel J, Lane R. A six-month double-blind, randomized, placebo-controlled study of a
transdermal patch in Alzheimer’s disease—rivastigmine patch versus capsule. Int J Geriatr
Psychiatry. 2007;22(5):456–67.
45. Doody RS, Geldmacher DS, Gordon B, Perdomo CA, Pratt RD, Donepezil Study Group.
Open-label, multicenter, phase 3 extension study of the safety and efficacy of donepezil in
patients with Alzheimer disease. Arch Neurol. 2001;58(3):427–33.
46. Winblad B, Engedal K, Soininen H, Verhey F, Waldemar G, Wimo A, Wetterholm AL,
Zhang R, Haglund A, Subbiah P, Donepezil Nordic Study Group. A 1-year, randomized,
placebo-controlled study of donepezil in patients with mild to moderate AD. Neurology.
2001;57(3):489–95.
47. Lin JS, O’Connor E, Rossom RC, Perdue LA, Eckstrom E. Screening for cognitive impair-
ment in older adults: a systematic review for the U.S. Preventive Services Task Force. Ann
Intern Med. 2013;159(9):601–12. Review.
48. US Department of Health and Human Services, Assistant Secretary for Planning and
Evaluation, Office of Disability, Aging and Long-Term Care Policy. Examining models of
dementia care: final report. 9 2016.
49. Epperly T, Dunay MA, Boice JL. Alzheimer disease: pharmacologic and nonpharmacologic
therapies for cognitive and functional symptoms. Am Fam Physician. 2017;95(12):771–8.
286 F. Trotta et al.
50. Molony SL, Kolanowski A, Van Haitsma K, Rooney KE. Person-centered assessment and care
planning. Gerontologist. 2018;58(suppl_1):S32–47.
51. McShane R, Westby MJ, Roberts E, Minakaran N, Schneider L, Farrimond LE, Maayan N, Ware
J, Debarros J. Memantine for dementia. Cochrane Database Syst Rev. 2019;3(3):CD003154.
52. Irvine, C.A. Namzaric (memantine and donepezil hydrochlorides). Full prescribing informa-
tion, Allergan USA, Inc. 2016.
53. FDA Grants Accelerated Approval for Alzheimer’s Drug. US Food and Drug Administration.
https://www.fda.gov/news-events/press-announcements/fda-grants-accelerated-approval-
alzheimers-drug. Accessed 7 June 2021.
54. McCleery J, Quinn TJ. Aducanumab and the certainty of evidence. Age Ageing.
2021;50(6):1899–900.
55. Cummings J, Salloway S. Aducanumab: appropriate use recommendations. Alzheimers
Dement. 2021;18(3):531–3.
56. Aduhelm (Aducanumab-AVWA injection). US FDA approved product information.
Cambridge: Biogen Inc; 2021.
57. Droś J, Klimkowicz-Mrowiec A. Current view on post-stroke dementia. Psychogeriatrics.
2021;21(3):407–17.
58. Mijajlović MD, Pavlović A, Brainin M, Heiss WD, Quinn TJ, Ihle-Hansen HB, Hermann
DM, Assayag EB, Richard E, Thiel A, Kliper E, Shin YI, Kim YH, Choi S, Jung S, Lee YB,
Sinanović O, Levine DA, Schlesinger I, Mead G, Milošević V, Leys D, Hagberg G, Ursin
MH, Teuschl Y, Prokopenko S, Mozheyko E, Bezdenezhnykh A, Matz K, Aleksić V, Muresanu
D, Korczyn AD, Bornstein NM. Post-stroke dementia—a comprehensive review. BMC Med.
2017;15(1):11.
59. Leys D, Hénon H, Mackowiak-Cordoliani MA, Pasquier F. Poststroke dementia. Lancet
Neurol. 2005;4(11):752–9.
60. Williamson JD, Supiano MA, Applegate WB, Berlowitz DR, Campbell RC, Chertow GM,
Fine LJ, Haley WE, Hawfield AT, Ix JH, Kitzman DW, Kostis JB, Krousel-Wood MA, Launer
LJ, Oparil S, Rodriguez CJ, Roumie CL, Shorr RI, Sink KM, Wadley VG, Whelton PK,
Whittle J, Woolard NF, Wright JT Jr, Pajewski NM, SPRINT Research Group. Intensive vs
standard blood pressure control and cardiovascular disease outcomes in adults aged ≥75 years:
a randomized clinical trial. JAMA. 2016;315(24):2673.
61. Bavishi C, Bangalore S, Messerli FH. Outcomes of intensive blood pressure lowering in older
hypertensive patients. J Am Coll Cardiol. 2017;69(5):486.
62. Corvol JC, Bouzamondo A, Sirol M, Hulot JS, Sanchez P, Lechat P. Differential effects of
lipid-lowering therapies on stroke prevention: a meta-analysis of randomized trials. Arch
Intern Med. 2003;163(6):669.
63. Oesterle A, Laufs U, Liao JK. Pleiotropic effects of statins on the cardiovascular system. Circ
Res. 2017;120(1):229.
64. Kernan WN, Ovbiagele B, Black HR, Bravata DM, Chimowitz MI, Ezekowitz MD, Fang
MC, Fisher M, Furie KL, Heck DV, Johnston SC, Kasner SE, Kittner SJ, Mitchell PH, Rich
MW, Richardson D, Schwamm LH, Wilson JA, American Heart Association Stroke Council,
Council on Cardiovascular and Stroke Nursing, Council on Clinical Cardiology, and Council
on Peripheral Vascular Disease. Guidelines for the prevention of stroke in patients with stroke
and transient ischemic attack: a guideline for healthcare professionals from the American
Heart Association/American Stroke Association. Stroke. 2014;45(7):2160.
65. Smith EE, Cieslak A, Barber P, Chen J, Chen YW, Donnini I, Edwards JD, Frayne R, Field
TS, Hegedus J, Hanganu V, Ismail Z, Kanji J, Nakajima M, Noor R, Peca S, Sahlas D, Sharma
M, Sposato LA, Swartz RH, Zerna C, Black SE, Hachinski V. Therapeutic strategies and drug
development for vascular cognitive impairment. J Am Heart Assoc. 2017;6(5):e005568.
66. Cummings J, Ritter A, Rothemberg K. Advances in management of neuropsychiatric syn-
dromes in neurodegenerative diseases. Curr Psychiatry Rep. 2019;21:79.
67. Tible OP, Riese F, Savaskan E, von Gunten A. Best practice in the management of behavioural
and psychological symptoms of dementia. Ther Adv Neurol Disord. 2017;10(8):297–309.
20 Pharmacological Treatment of Cognitive and Behavioral Disorders in Dementia 287
68. Eissa N, Sadeq A, Sasse A, Sadek B. Role of neuroinflammation in autism spectrum disorder
and the emergence of brain histaminergic system. Lessons also for BPSD? Front Pharmacol.
2020;11:886.
69. Calsolaro V, Femminella GD, Rogani S, Esposito S, Franchi R, Okoye C, Rengo G, Monzani
F. Behavioral and psychological symptoms in dementia (BPSD) and the use of antipsychotics.
Pharmaceuticals. 2021;14(3):246.
70. Garre-Olmo J, López-Pousa S, Vilalta-Franch J, de Gracia Blanco M, Vilarrasa AB. Grouping
and trajectories of the neuropsychiatric symptoms in patients with Alzheimer’s disease, part I:
symptom clusters. J Alzheimers Dis. 2010;22(4):1157–67.
71. Livingston G, Kelly L, Lewis-Holmes E, Baio G, Morris S, Patel N, Omar RZ, Katona C,
Cooper C. Non-pharmacological interventions for agitation in dementia: systematic review of
randomised controlled trials. Br J Psychiatry. 2014;205(6):436–42.
72. Reus VI, Fochtmann LJ, Eyler AE, Hilty DM, Horvitz-Lennon M, Jibson MD, Lopez OL,
Mahoney J, Pasic J, Tan ZS, Wills CD, Rhoads R, Yageret J. The American Psychiatric
Association practice guideline on the use of antipsychotics to treat agitation or psychosis in
patients with dementia. Am J Psychiatry. 2016;173(5):543–6.
73. Markota M, Rummans TA, Bostwick JM, Lapid MI. Benzodiazepine use in older adults: dan-
gers, management, and alternative therapies. Mayo Clin Proc. 2016;91(11):1632–9.
74. Brücke T, Roth J, Podreka I, Strobl R, Wenger S, Asenbaum S. Striatal dopamine D2-receptor
blockade by typical and atypical neuroleptics. Lancet. 1992;339(8791):497.
75. Faay MDM, Czobor P, Sommer IEC. Efficacy of typical and atypical antipsychotic medica-
tion on hostility in patients with psychosis-spectrum disorders: a review and meta-analysis.
Neuropsychopharmacology. 2018;43(12):2340–9.
76. Aringhieri S, Carli M, Kolachalam S, Verdesca V, Cini E, Rossi M, McCormick PJ, Corsini
GU, Maggio R, Scarselli M. Molecular targets of atypical antipsychotics: from mechanism of
action to clinical differences. Pharmacol Ther. 2018;192:20–41.
77. Mintzer JE, Tune LE, Breder CD, Swanink R, Marcus RN, McQuade RD, Forbes
A. Aripiprazole for the treatment of psychoses in institutionalized patients with Alzheimer
dementia: a multicenter, randomized, double-blind, placebo-controlled assessment of three
fixed doses. Am J Geriatr Psychiatry. 2007;15(11):918–31.
78. De Deyn PP, Drenth AF, Kremer BP, Oude Voshaar RC, Van Dam D. Aripiprazole in the treat-
ment of Alzheimer’s disease. Expert Opin Pharmacother. 2013;14(4):459–74.
79. Schneider LS, Tariot PN, Dagerman KS, Davis SM, Hsiao JK, Ismail MS, Lebowitz BD,
Lyketsos CG, Ryan JM, Stroup TS, Sultzer DL, Weintraub D, Lieberman JA, CATIE-AD
Study Group. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer’s dis-
ease. N Engl J Med. 2006;355(15):1525–38.
80. Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psycho-
logical symptoms of dementia. BMJ. 2015;350:h369.
81. Tampi RR, Tampi DJ, Balachandran S, Srinivasan S. Antipsychotic use in dementia: a system-
atic review of benefits and risks from meta-analyses. Ther Adv Chronic Dis. 2016;7:229–45.
Pharmacological Treatment
of Osteoporosis in Older Patients 21
Marian Dejaeger, Jolan Dupont, Michaël R. Laurent,
and Evelien Gielen
Osteoporosis is a systemic bone disease, characterized by low bone mass and dis-
ruptions in the micro-architectural structure of bone tissue, resulting in an increase
in bone fragility and fracture risk [1]. The incidence of osteoporotic fractures
increases with age, and osteoporosis at advanced age is a challenge because of coex-
isting comorbidities and the major post-fracture implications in terms of morbidity,
loss of mobility, and mortality. Currently, the cumulative incidence of hip and ver-
tebral fractures in women at the age of 80 years is close to 30% and more than 40%,
respectively. Women over the age of 80 represent more than 30% of all osteoporotic
fractures and even more than 60% of all non-vertebral fractures [2–4]. Age is also
one of the main determinants of the type of osteoporotic fracture. Between the ages
of 55 and 75, the risk of vertebral fractures peaks in women, whereas beyond the
age of 75, women become increasingly at risk of non-vertebral fractures, e.g., hip
fractures [5]. The burden of osteoporosis will only increase further because of the
aging society.
Osteoporosis and osteoporotic fractures tend to occur in a part of the older popula-
tion already suffering from frailty [6]. Indeed, older persons with osteoporotic frac-
tures are not “average” older persons but frail individuals with a high prevalence of
underlying comorbidities and at risk of disabilities [7]. This frailty is reflected in the
poor post-fracture outcomes of these patients. For example, 1 year post-fracture,
40% of the patients are still unable to walk independently, 33% are institutionalized
or totally dependent, and 20% are deceased [8, 9]. These poor outcomes are not
restricted to hip fractures alone, as similar observations have been made in older
persons with other types of non-vertebral fractures and even vertebral fractures [10].
Moreover, the excess mortality is not only observed within the first year after the
fracture but persists in the years thereafter due to the underlying comorbidities of
these frail older persons [11, 12].
Despite the high prevalence and significant burden of osteoporosis, this condition
remains to be underdiagnosed and undertreated. This is most prominent in women
aged ≥80 years [13]. To illustrate, according to a study in more than 700,000 per-
sons aged ≥75 years in Sweden, 16.1% of women and 3.4% of men used osteopo-
rosis medication, lower than the estimated prevalence of osteoporosis in that
country. In particular in persons ≥90 years, osteoporosis was largely under-
treated [14].
Yet, the operational definition of osteoporosis also applies to older persons. The
World Health Organization (WHO) defines osteoporosis as a bone mineral density
(BMD) value at the lumbar spine or the proximal femur of at least −2.5 standard
deviations (SD) below the peak bone mass of a young healthy individual [15].
However, sensitivity of dual-energy X-ray absorptiometry (DXA) for fracture is
low, as the majority of fractures occurs in persons with a T-score above −2.5. This
implies that factors other than BMD contribute to fracture risk [16]. More specifi-
cally, an existing osteoporotic fracture is the most significant predictor of future
fracture risk, resulting in a doubling of fracture risk compared to persons without
previous fractures. Also, increasing age itself is an important risk factor for fractures
as over a 4 SD interval of BMD (+1 to −3 SD), a 14-fold increase in hip fracture risk
is seen at the age of 50, but a 145-fold increase at the age of 80 [17, 18]. Therefore,
fracture risk assessment tools based on age and other clinical risk factors are increas-
ingly being used to target osteoporosis therapy. Among these is the FRAX algo-
rithm, which includes the weight of various clinical risk factors for fractures, with
or without femoral neck BMD, to estimate the 10-year probability of hip fracture
and major osteoporotic fractures (clinical spine, forearm, hip, or shoulder fractures)
[19]. The main goal of FRAX is to identify individuals at higher fracture risk who
qualify for osteoporosis treatment. Three approaches can be distinguished, which,
21 Pharmacological Treatment of Osteoporosis in Older Patients 291
especially in older persons, differ in the threshold above which treatment is recom-
mended: fixed, age-dependent, and hybrid thresholds.
First, according to the fixed model of the USA National Osteoporosis Foundation
(NOF), treatment should be initiated when the 10-year fracture risk of a person
exceeds the 10-year fracture probability at which treatment becomes cost-effective
[20, 21]. Second, the age-dependent model of the UK National Osteoporosis
Guideline Group (NOGG) indicates that treatment should be considered when the
fracture probability of an individual exceeds the FRAX intervention threshold,
defined as the age-specific fracture probability of a man or woman with a previous
fragility fracture, no other risk factors, an average BMI (24 kg/m2), and without
knowledge of BMD [22, 23]. The rationale is that in general, a person with a previ-
ous fragility fracture is considered to qualify for treatment. Third, as the previous
model induces inequalities in access to therapy especially at older age, an alterna-
tive threshold using a hybrid model has been proposed by the NOGG. According to
this model, the intervention threshold rises with age but remains constant from the
age of 70, being at 20% for a major osteoporotic fracture and 5.4% for a hip fracture
[23, 24].
In this section, we will discuss the efficacy and safety of the pharmacological treat-
ment options for osteoporosis in older persons. Osteoporosis treatment for older
persons should be proven to be effective in this cohort, not only against vertebral
fractures but even more so against non-vertebral fractures, as these account for the
majority of negative outcomes (e.g., morbidity, mortality). Treatment should also be
safe in frail older adults, with underlying comorbidities and at increased risk of
adverse events.
One of the main determinants of bone loss and fracture risk in old age is calcium
and vitamin D deficiency. Therefore, combined supplementation of calcium and
vitamin D has become one of the main treatment strategies for reducing bone loss
and fracture risk in old age. Low vitamin D status and insufficient dietary calcium
intake can lead to secretion of parathyroid hormone (age-related hyperparathyroid-
ism), stimulating bone resorption and increasing the fracture risk.
Despite a high degree of consensus on the essential role of vitamin D for bone
health and other extra-skeletal effects, there remains some lack of consensus regard-
ing the optimal concentration of total 25-hydroxyvitamin D (25(OH)D). Yet, it is
clear that vitamin D insufficiency occurs in all age groups. In fact, a gradual decline
in 25(OH)D levels from healthy adults over independent older adults to institution-
alized individuals and hip fracture patients can be observed [25]. Severe vitamin D
deficiency (<10 μg/L) is seen in almost 4 out of 5 institutionalized people [25]. The
292 M. Dejaeger et al.
Alendronate
The efficacy of alendronate in postmenopausal women was demonstrated by the
Fracture Intervention Trial (FIT). The FIT Vertebral Fracture Arm (FIT I) included
women who suffered from vertebral fracture(s), while the FIT Clinical Fracture
Arm (FIT II) included women with a T-score of ≤−1.6 at the femoral neck (mean
age 70.8 years and 67.7 years, respectively) [39, 40].
Two sub-analyses of the FIT trials have been undertaken to determine the anti-
fracture efficacy and safety of alendronate in older persons. A post hoc analysis of
FIT I has focused on the efficacy of alendronate in postmenopausal women with the
highest fracture risk, including a subgroup of women aged ≥75 years (range
75–82 years) [41]. In order to prevent one new vertebral fracture, eight women aged
≥75 years (RR = 0.62; 95% CI, 0.41–0.94) needed to be treated for 3 years with
alendronate compared with nine women aged <75 years (RR = 0.49; 95% CI,
0.35–0.68). Another analysis, based on pooled data from both FIT arms, aimed to
calculate age-specific fracture rates by treatment group (55 to <65 years, 65 to
<70 years, 70 to <75 years, and 75–85 years) [42]. Relative risk reductions were
comparable among age groups for hip (RR = 0.47; 95% CI, 0.27–0.81; p < 0.01) and
vertebral (RR = 0.55; 95% CI, 0.37–0.83; p < 0.01) fractures, revealing an even
greater absolute risk reduction as age increases, which can be explained by the age-
related increase in fracture risk in the placebo group [42]. A study on older patients
(range 71–92 years) with a previous fracture showed also a significant reduction in
hip fracture risk (hazard ratio (HR) = 0.72; 95% CI, 0.61–0.85, p < 0.001), which
was maintained across all age quartiles [43]. Finally, using a national database of
men and women undergoing a fall risk assessment at a Swedish healthcare facility,
the efficacy of alendronate treatment was investigated in patients aged ≥80 years
(mean age of 85.7+/− 3.9 years) and history of fracture [43]. Alendronate was asso-
ciated with a lower hip fracture risk (HR = 0.66; 95% CI, 0.51–0.86; P < 0.01).
The post hoc analysis of FIT I and the pooled analysis from both FIT arms did
not report safety data in older persons [41, 42]. The Swedish study reported a
reduced mortality risk in patients treated with alendronate (HR = 0.88; 95% CI,
Table 21.1 Relative risk (95% CI) of new vertebral, hip and non-vertebral fractures compared with placebo in older women receiving currently available
294
osteoporosis treatments
Mean age
RCT Included participants N (years) Vertebral fractures Hip fractures Non-vertebral fractures
Alendronate Post hoc analysis Women aged 539 Not RR = 0.62; – –
FIT I (3 years) 75–82 y specified 95% CI 0.41–0.94.
(Ensrud 1997) p < 0.05
pinteract < 75 & ≥75 y NS
Pooled analysis Women aged 3658 RR = 0.55; RR = 0.47; –
FIT I and FIT II 55–80 y 95% CI 0.37–0.83 95% CI 0.27–0.81
(3–4 years) 55–<65 y (constant RR) (constant RR)
(Hochberg 2005) 65–<70 y
70–<75 y
75–85 y
Axelsson et al. Women aged 110.190 82.4 y – HR = 0.72; –
(5 years) 71.1–92.3 y with a 95% CI: 0.61–0.85;
(Axelsson 2016) prior fracture p < 0.001
Risedronate HIP - arm 2 Women aged ≥80 y 3886 83 y – RR = 0.8; 10.8% (Risedronate) vs
(3 years) with at least one 95% CI 0.6–1.2; 11.9% (placebo);
(McClung 2001) non-skeletal risk p = 0.35 p = 0.43
factor for hip
fracture
or T-score at
FN < −4 or
<−3 + hip axis
length of ≥11.1 cm
Post hoc pooled Women aged ≥80 y 1392 83 y HR = 0.56; – 14.0% (Risedronate) vs
analysis VERT-NA, with T-score <−2.5 95% CI 0.39–0.81; 16.2% (placebo);
VERT-MN and HIP at FN or at least one p = 0.003 p = 0.66
(3 years) prevalent vertebral pinteract < 80 & ≥85 y NS
(Boonen 2004b) fracture
M. Dejaeger et al.
Mean age
RCT Included participants N (years) Vertebral fractures Hip fractures Non-vertebral fractures
21
Zoledronic Post hoc analysis Women aged ≥75 y 3888 79.4 y HR = 0.34; HR = 0.82; HR = 0.73;
acid HORIZON-PFT with T-score ≤−2.5 95% CI 0.21–0.55; 95% CI 0.56–1.2; 95% CI 0.60–0.90;
and RFT (3 years) at FN or ≥1 vertebral p < 0.001 p = 0.297 p = 0.002
(Boonen 2010) or hip fracture pinteract < 75 & ≥75 y NS pinteract < 70 & ≥75 y SS pinteract < 70 & ≥75 y NS
Denosumab Post hoc analysis Women aged ≥75 y 2471 78.2 y – 0.9% (Denosumab) –
FREEDOM vs 2.3% (placebo);
(3 years) (Boonen p < 0.01; ARR 1.4%
2011) pinteract < 75 & ≥75 y NS
Preplanned analysis Women aged ≥75 y 2471 78.2 y 3.1% (Denosumab) vs – 7.9% (Denosumab) vs
FREEDOM 8.6% (placebo); 9.0% (placebo);
(3 years) (McClung RR = 0.36; RR = 0.84;
2012) 95% CI 0.25–0.53 95% CI 0.63–1.12
pinteract < 75 & ≥75 y NS pinteract < 75 & ≥75 y NS
FREEEDOM Women aged ≥75 y 662 78 y 3.6% (Denosumab) 1.0% (Denosumab) 5.4% (Denosumab)
Extension (6 years) (no placebo group) (no placebo group) (no placebo group)
(Papapoulos 2012)
Teriparatide Prespecified Women aged ≥75 y 244 78.3 y RR = 0.35, – RR = 0.75; p = 0.661
subgroup analysis p < 0.05 (not powered)
FPT (19 months) pinteract < 75 & ≥75 y NS pinteract < 75 & ≥75 y NS
(Boonen 2006a)
Abalopratide Posthoc analysis Women aged ≥80 y 94 81.7 0 fractures 1 fracture
ACTIVE (Aboloparatide) vs 2 (Abaloparatide) vs 2
(18 months) fractures (placebo) fractures (placebo)
Pharmacological Treatment of Osteoporosis in Older Patients
McClung 2018
Posthoc analysis Women aged ≥75 y 303 NA <65 y, 65–75 y, ≥75 y: <65 y, 65–75 y, ≥75 y:
ACTIVE consistent fracture risk consistent fracture risk
(18 months) reduction reduction
Cosman 2017
(continued)
295
Table 21.1 (continued)
296
Mean age
RCT Included participants N (years) Vertebral fractures Hip fractures Non-vertebral fractures
Romosozumab FRAME (2 years) Women 55–90 y 7180 70.9 y 12 months (Romo vs 12 months (Romo vs 12 months (Romo vs
Cosman 2016 placebo): RR = 0.27; placebo): HR = 0.54; placebo): HR = 0.75;
95% CI 0.16–0.47 95% CI 0.22–1.35 95% CI 0.53–1.05
24 months (Romo/ 24 months (Romo/ 24 months (Romo/
Dmab vs placebo/ Dmab vs placebo/ Dmab vs placebo/
Dmab): RR = 0.25; Dmab): HR = 0.50; Dmab): HR = 0.75;
95% CI 0.16–0.40 95% CI 0.24–1.04 95% CI 0.57–0.97
ARCH (2 years) Women 55–90 y 4093 74.3 y 12 months (Romo vs 12 months (Romo vs 12 months (Romo vs
Saag 2017 ALN): RR = 0.63; ALN): HR = 0.64; ALN): HR = 0.74;
95% CI 0.47–0.85 95% 0.33–1.26 95% CI 0.54–1.01
24 months (Romo/ Primary analysis Primary analysis
ALN vs ALN/ALN): (Romo/ALN vs ALN/ (Romo/ALN vs ALN/
RR = 0.52; 95% CI ALN): HR = 0.62; ALN): HR = 0.81;
0.40–0.66 95% CI 0.42–0.92 95% CI 0.66–0.99
FN Femoral neck, LS Lumbar spine, y Years, ITT Intention to treat, NS Not significant, RR Risk ratio, HR Hazard ratio, Romo Romosozumab, Dmab Denosumab,
ALN Alendronate. Results in bold indicate significant results
Table adapted from: Vandenbroucke A et al. Pharmacological treatment of osteoporosis in the oldest old. Clinical Interventions in Aging 2017:12; 1065–1077.
Dove Medical Press Limited
M. Dejaeger et al.
21 Pharmacological Treatment of Osteoporosis in Older Patients 297
Risedronate
The Vertebral Efficacy with Risedronate Therapy North America (VERT-NA) and
Multinational (VERT-MN) studies showed both that risedronate can efficiently
reduce vertebral and non-vertebral fractures rates [44, 45] in postmenopausal
women with one or more vertebral fractures (mean age +/− 70 years) [44, 45]. In
addition, the HIP trial has studied the effect of risedronate on hip fracture risk in two
different arms, of which the first arm included postmenopausal women with a mean
age of 74 years [46]. To determine the efficacy of risedronate in older persons, the
second arm of the HIP trial included 3886 women, aged ≥80 years (mean age
83 years). These older women were included based on having a low BMD at the
femoral neck or at least one nonskeletal risk factor for hip fracture (e.g., poor gait or
a propensity to fall) [46]. After 3 years of risedronate use, no significant reduction
in the risk of hip fractures was observed (RR = 0.8; 95% CI, 0.6–1.2; p = 0.35).
Only in the subgroup of women (16%) included based on low BMD, a significant
reduction in hip fracture rate was seen. However, most of the participants (58%)
were included on the basis of a nonskeletal risk factor. A second analysis on pooled
data from these three studies (VERT-NA, VERT-MN, and HIP trials) focused on
older individuals [47]. This analysis included 1392 women aged ≥80 years (mean
age 83 years) with osteoporosis (T-score <−2.5 or at least one vertebral fracture).
After 1 year, the vertebral fracture risk decreased by 81% (HR = 0.19; 95% CI,
0.09–0.40; P < 0.001) and after 3 years by 44% (HR = 0.56; 95% CI, 0.39–0.81;
p = 0.003). Regarding non-vertebral fractures, the incidence did not differ signifi-
cantly between the treatment group and the placebo group (p = 0.66).
This gap in benefit for vertebral vs non-vertebral fractures in both analyses in
older persons might be explained by the knowledge that bisphosphonates impact
on BMD, demonstrated by the significant reduction of vertebral fractures [46, 47].
In spite of this, bisphosphonates do not intervene with the nonskeletal risk factors
of fractures such as gait disturbances, impaired balance, and fall risk. These non-
skeletal factors play a major role in the occurrence of non-vertebral fractures,
such as hip fractures, in older persons, who are more at risk of falling [48]. On the
contrary, vertebral fractures are often spontaneous or atraumatic, thus less influ-
enced by these nonskeletal risk factors. Furthermore, the discrepancy between the
older and younger population in preventing non-vertebral fractures might be
explained, at least partly, due to insufficient statistical power in the older age
group [47].
298 M. Dejaeger et al.
Older women (aged ≥80 years) in the HIP trial had a marginally higher inci-
dence of death, other serious adverse events, and discontinuation due to adverse
events than younger women [46]. However, the overall occurrence and types of
adverse events, including those involving the upper gastrointestinal tract, were com-
parable in the risedronate and placebo group, aside from age. Additionally, in
women aged ≥80 years in the pooled analysis of the VERT-NA, VERT-MN, and
HIP trials, adverse events were similar in the risedronate and placebo group [47].
Even in older persons with active gastrointestinal tract disease at baseline or on
aspirin, nonsteroidal anti-inflammatory drugs (NSAIDs), or proton pump inhibitors
(PPIs), the incidence of adverse events was similar in both treatment groups.
Thus, safety data of oral bisphosphonates confirm that they are well tolerated in
older persons and not associated with an increased risk of adverse events as com-
pared to placebo. Nonetheless, in clinical practice, treatment with oral bisphospho-
nates may be challenging, as the rather demanding intake instructions (e.g., to stay
upright for at least 30 min after the intake) may be difficult for physically or cogni-
tively impaired persons. Inappropriate administration, however, increases the risk of
adverse events and potentially further decreases the already low oral bioavailability
(around 1%–2%) [38, 49, 50]. Furthermore, oral osteoporosis medication rises the
pill burden in older persons, which contributes to low compliance. Indeed, almost
50% of those who have been prescribed an oral bisphosphonate have discontinued
treatment after 1 year [51].
Zoledronic Acid
Usage of an intravenous bisphosphonate, such as zoledronic acid, might provide an
alternative in older persons that cannot tolerate or adhere to oral bisphosphonates
[52]. The Health Outcomes and Reduced Incidence with Zoledronic Acid Once
Yearly Pivotal Fracture Trial (HORIZON-PFT) demonstrated that once-yearly infu-
sion of zoledronic acid significantly reduces the risk of vertebral, hip, and non-
vertebral fractures in postmenopausal women with osteoporosis (mean age 73 years)
[53]. In a second analysis in participants with surgical repair of a low-trauma hip
fracture (mean age 74.4 years), the HORIZON-Recurrent Fracture Trial (HORIZON-
RFT), zoledronic acid significantly lowers the risk of new vertebral and new non-
vertebral fractures [54].
Additional post hoc pooled analysis of the HORIZON-PFT and HORIZON-RFT
was performed, focusing on older persons, by including postmenopausal women
aged ≥75 years (mean age 79.4 years) with osteoporosis (T-score ≤−2.5 at the
femoral neck or ≥1 prevalent vertebral or hip fracture) [52]. After 3 years of treat-
ment, incidence of any clinical vertebral (1.1% vs 3.7%) and non-vertebral fracture
(9.9% vs 13.7%) was significantly lower in the zoledronic acid group compared to
the placebo group (HR = 0.34; 95% CI 0.21–0.55; p < 0.001 and HR = 0.73; 95%
CI 0.6–0.9; p = 0.002 respectively). This benefit was similar to the relative risk
reduction in subjects younger than 75 years in the HORIZON-PFT and HORIZON-
RFT. Hence, zoledronic acid is an efficient treatment option for the prevention of
vertebral and non-vertebral fractures in older persons. However, in patients aged
≥75 years, the incidence of hip fractures decreased with zoledronic acid, but this did
21 Pharmacological Treatment of Osteoporosis in Older Patients 299
Denosumab
Denosumab is a human monoclonal antibody that acts on RANKL, a mediator of
bone resorption. In the Fracture Reduction Evaluation of Denosumab in
Osteoporosis Every 6 months (FREEDOM) trial, 7800 women between 60 and
90 years (mean age 72.3 years) with a BMD T-score of <−2.5 but not <−4.0 at the
lumbar spine or total hip received denosumab or placebo for 3 years. Denosumab
was linked with a significant reduction in the risk of vertebral, hip, and non-verte-
bral fractures [55].
An additional post hoc analysis of the FREEDOM trial focused on the effect of
denosumab in populations at high risk of fractures, among which women aged
≥75 years [56]. In this age group (mean age 78.2 years), denosumab lowered the
risk of hip fractures by 62% (2.3% placebo vs 0.9% denosumab; p < 0.01). This risk
reduction in older patients is similar with the risk reduction in the overall study
population of the FREEDOM trial. One more analysis of the FREEDOM trial con-
firmed that denosumab lowers the risk of vertebral fractures to the same extent in
subjects aged ≥75 years (RR = 0.36; 95% CI, 0.25–0.53) as in subjects <75 years
(RR = 0.30; 95% CI, 0.22–0.41; interaction p-value, 0.482) [57]. Moreover, the
effect on non-vertebral fractures in persons older than 75 years (RR = 0.84; 95% CI,
0.63–1.12) was comparable to younger persons (RR = 0.78; 95% CI, 0.63–0.96;
interaction p-value, 0.642). Also, the FREEDOM Extension trial demonstrated that
also in women aged ≥75 years, denosumab for up to 6 years was associated with a
persistently low incidence of new vertebral, hip, and non-vertebral fractures, with a
fracture incidence similar to that observed after 3 years [58].
Hence, denosumab is effective in preventing vertebral and hip fractures in older
persons [56, 57]. In contrast, as indicated, no significant reduction in hip fracture
risk was shown for the bisphosphonates risedronate and zoledronic acid in older
persons, although, as mentioned, this might be explained by lack of statistical power
[41, 42, 52]. However, it is tempting to speculate that this observation of hip fracture
reduction with denosumab is due to the mechanism of action of denosumab, differ-
ent from that of bisphosphonates, with distinct effects on cortical bone [59]. Cortical
porosity is indeed one of the main determinants of non-vertebral fracture risk,
including hip fracture risk.
300 M. Dejaeger et al.
Teriparatide
In the Fracture Prevention Trial (FPT), teriparatide lowered the risk of vertebral
fractures by 65% (HR = 0.35; 95% CI, 0.22–0.55; P < 0.001) and the risk of non-
vertebral fractures by 53% (HR = 0.47; 95% CI, 0.25–0.88; P = 0.04) in postmeno-
pausal women with a prior vertebral fracture (mean age 69.5 years) [71]. In a
subgroup analysis on the FPT study, Boonen et al. investigated the effect of teripa-
ratide in persons aged ≥75 years after 19 months of treatment (mean age 78.3 years)
[72]. About 5.2% of the older persons in the teriparatide group and 15.1% in the
placebo group suffered a new vertebral fracture (RR = 0.35, P < 0.05). Hip fracture
incidence was not a primary endpoint in this trial. Treatment-by-age interaction was
not significant (p = 0.99), indicating that there was no significant difference in the
effect of teriparatide in younger vs older patients. Also in the older subgroup, 3.2%
of the women in the teriparatide group and 4.2% in the placebo group had a new
non-vertebral fracture (RR = 0.75; p = 0.661). Again, the treatment-by-age interac-
tion was not significant (p = 0.42). The nonsignificant effect on non-vertebral frac-
ture risk in older persons can be justified by the small number of non-vertebral
fractures in the older subgroup, resulting in a lack of power for this aspect [72]. In
conclusion, teriparatide has a significant effect on vertebral and non-vertebral frac-
ture risk in young as well as in older fracture patients.
In the post hoc analysis of the FPT trial in women aged ≥75 years, there was no
rise in adverse events in women treated with teriparatide compared to placebo [72].
On the contrary, back pain, cataract, and pruritus were significantly less present in
those treated with teriparatide. Treatment-by-age interaction (≥75 vs <75 years)
was not significant for the major adverse events. Cataract, deafness, pruritus, and
weight loss were reported to be less frequent in the older compared to the younger
age group, whereas only diarrhea was reported to be more frequent. Thus, in older
persons, the safety profile of teriparatide is comparable to placebo. In clinical prac-
tice, the major disadvantages of teriparatide are the cost and the daily subcutaneous
administration which may be a hamper for older patients.
Abaloparatide
The pivotal phase 3 trial ACTIVE (Abaloparatide Comparator Trial In Vertebral
Endpoints) and its extension (ACTIVExtend) showed that 18 months of abalopara-
tide, a 34-amino acid PTH-related peptide (PTHrP), followed by sequential treat-
ment with alendronate for an additional 24 months, significantly reduced the risk of
vertebral, non-vertebral, clinical, and major osteoporotic fractures in postmeno-
pausal women with osteoporosis (mean age of 68.8 years) [73].
Two post hoc analyses, by McClung et al. and Cosman et al., investigated the
efficacy and safety of abaloparatide in women aged ≥80 years [74] and ≥75 years
[75], respectively. In the analysis of McClung et al., abaloparatide was associated
302 M. Dejaeger et al.
with numerical, but not statistically significant reductions in the risk of vertebral
and non-vertebral fractures (two vertebral and two non-vertebral fractures in the
placebo group vs. zero and one fracture, respectively, in the abaloparatide group)
[74]. These numerical reductions in fracture risk are consistent with the effective-
ness of abaloparatide in the overall ACTIVE population, but the difference is not
statistically significant, which may be explained by the very small number of frac-
tures in the older age group [74]. In the analysis by Cosman et al., the effect of
abaloparatide on vertebral and non-vertebral fracture risk reduction was consistent
across different age groups (<65 vs. 65 to <75 vs. ≥75 years) [75]. In these separate
age groups, vertebral and non-vertebral fracture risk reduction was not significant,
which may be explained by the small number of participants and events in these
subgroups.
Regarding tolerability and safety, the proportion of women reporting adverse
events was similar in the older age group (≥80 years) and the overall ACTIVE
population [74]. However, the older patients reported more serious adverse events
than the overall ACTIVE population. This may not be unexpected for an older popu-
lation but, more importantly, did not differ between abaloparatide and placebo [74].
Romosozumab
In the FRAME (Fracture Study in Postmenopausal Women with Osteoporosis)
study in postmenopausal women aged 55–90 years with osteoporosis, romoso-
zumab was associated with a lower risk of vertebral fractures than placebo at
12 months and, after the transition to denosumab, at 24 and 36 months [76, 77].
At 12 months, the risk of clinical fracture was also significantly lower with romo-
sozumab than with placebo. The ARCH study (Active-Controlled Fracture Study
in Postmenopausal Women with Osteoporosis at High Risk) showed that in post-
menopausal women aged 55 to 90 years at high risk of fracture, 12 months of
romosozumab followed by alendronate resulted in a significantly lower risk of
clinical, vertebral, non-vertebral, and hip fractures compared to alendronate
alone [78].
No post hoc analyses have been performed regarding the efficacy of romoso-
zumab in older persons, but the mean age in FRAME and ARCH was 71 years and
74.3 years, respectively, with 31.2% and 52% of included women being ≥75 years.
sufficiently powered for vertebral fracture. This study showed that men treated with
zoledronic acid had fewer vertebral fractures than those on placebo [81]. The effect
of denosumab was investigated in the ADAMO trial in 242 men, showing a signifi-
cant increase of BMD at the spine and hip after 12 months of treatment vs placebo
[82]. The study was not powered for fractures. Teriparatide significantly increases
BMD in men with osteoporosis and is licensed by the FDA and EMA as treatment
for osteoporosis in men [83]. Romosozumab for 12 months increased BMD at the
spine and hip in men in the BRIDGE study, yet fracture endpoints were not included.
According to the authors, based on the observed BMD gains and similar fracture
risk, the fracture reduction observed in the FRAME study in postmenopausal
women can be extrapolated to men [84]. While in some countries (Japan, Australia,
etc.) romosozumab is approved for men at high risk of fractures, this is not the case
in Europe. Finally, testosterone replacement results in a small increase in BMD but
not as large as osteoporosis treatments and no data are available on fracture risk.
Thus, testosterone (taking also the potential side effects into account) is not consid-
ered as an adequate treatment option for osteoporosis in men.
Hence, we conclude that men seem to respond to the available osteoporosis treat-
ments in the same way as women, and therefore, the treatment approach, for the
most part, is similar in both genders.
21.5 Conclusion
References
1. Cosman F, de Beur SJ, LeBoff MS, Lewiecki EM, Tanner B, Randall S, et al. Clinician’s guide
to prevention and treatment of osteoporosis. Osteoporos Int. 2014;25(10):2359–81.
2. Cooper C, Campion G, Melton LJ 3rd. Hip fractures in the elderly: a world-wide projection.
Osteoporos Int. 1992;2(6):285–9.
3. Grados F, Marcelli C, Dargent-Molina P, Roux C, Vergnol JF, Meunier PJ, et al. Prevalence
of vertebral fractures in French women older than 75 years from the EPIDOS study. Bone.
2004;34(2):362–7.
4. Seeman E, Boonen S, Borgstrom F, Vellas B, Aquino JP, Semler J, et al. Five years treatment
with strontium ranelate reduces vertebral and nonvertebral fractures and increases the number
and quality of remaining life-years in women over 80 years of age. Bone. 2010;46(4):1038–42.
5. Sambrook P, Cooper C. Osteoporosis. Lancet. 2006;367(9527):2010–8.
6. Boonen S, Autier P, Barette M, Vanderschueren D, Lips P, Haentjens P. Functional outcome
and quality of life following hip fracture in elderly women: a prospective controlled study.
Osteoporos Int. 2004;15(2):87–94.
21 Pharmacological Treatment of Osteoporosis in Older Patients 305
7. Gielen E, Verschueren S, O'Neill TW, Pye SR, O'Connell MD, Lee DM, et al. Musculoskeletal
frailty: a geriatric syndrome at the core of fracture occurrence in older age. Calcif Tissue Int.
2012;91(3):161–77.
8. Leibson CL, Tosteson AN, Gabriel SE, Ransom JE, Melton LJ. Mortality, disability, and nurs-
ing home use for persons with and without hip fracture: a population-based study. J Am Geriatr
Soc. 2002;50(10):1644–50.
9. Schnell S, Friedman SM, Mendelson DA, Bingham KW, Kates SL. The 1-year mortal-
ity of patients treated in a hip fracture program for elders. Geriatr Orthop Surg Rehabil.
2010;1(1):6–14.
10. Gielen E, Vanderschueren D, Callewaert F, Boonen S. Osteoporosis in men. Best Pract Res
Clin Endocrinol Metab. 2011;25(2):321–35.
11. Haentjens P, Magaziner J, Colon-Emeric CS, Vanderschueren D, Milisen K, Velkeniers B,
et al. Meta-analysis: excess mortality after hip fracture among older women and men. Ann
Intern Med. 2010;152(6):380–90.
12. Tran T, Bliuc D, Hansen L, Abrahamsen B, van den Bergh J, Eisman JA, et al. Persistence
of excess mortality following individual nonhip fractures: a relative survival analysis. J Clin
Endocrinol Metab. 2018;103(9):3205–14.
13. Feldstein A, Elmer PJ, Orwoll E, Herson M, Hillier T. Bone mineral density measurement and
treatment for osteoporosis in older individuals with fractures: a gap in evidence-based practice
guideline implementation. Arch Intern Med. 2003;163(18):2165–72.
14. Johnell K, Fastbom J. Undertreatment of osteoporosis in the oldest old? A nationwide study of
over 700,000 older people. Arch Osteoporos. 2009;4(1–2):17–23.
15. Kanis JA, Melton LJ 3rd, Christiansen C, Johnston CC, Khaltaev N. The diagnosis of osteopo-
rosis. J Bone Miner Res. 1994;9(8):1137–41.
16. Thomas-John M, Codd MB, Manne S, Watts NB, Mongey AB. Risk factors for the devel-
opment of osteoporosis and osteoporotic fractures among older men. J Rheumatol.
2009;36(9):1947–52.
17. Melton LJ 3rd, Atkinson EJ, Cooper C, O'Fallon WM, Riggs BL. Vertebral fractures predict
subsequent fractures. Osteoporos Int. 1999;10(3):214–21.
18. Kanis JA, Johnell O, Oden A, Dawson A, De Laet C, Jonsson B. Ten year probabilities
of osteoporotic fractures according to BMD and diagnostic thresholds. Osteoporos Int.
2001;12(12):989–95.
19. FRAX®. Fracture Risk Assessment Tool. http://www.shef.ac.uk/FRAX/.
20. Kanis JA, McCloskey EV, Johansson H, Cooper C, Rizzoli R, Reginster JY, et al. European
guidance for the diagnosis and management of osteoporosis in postmenopausal women.
Osteoporos Int. 2013;24(1):23–57.
21. Tosteson AN, Melton LJ 3rd, Dawson-Hughes B, Baim S, Favus MJ, Khosla S, et al. Cost-
effective osteoporosis treatment thresholds: the United States perspective. Osteoporos Int.
2008;19(4):437–47.
22. Compston J, Bowring C, Cooper A, Cooper C, Davies C, Francis R, et al. Diagnosis and
management of osteoporosis in postmenopausal women and older men in the UK: National
Osteoporosis Guideline Group (NOGG) update 2013. Maturitas. 2013;75(4):392–6.
23. McCloskey E, Kanis JA, Johansson H, Harvey N, Oden A, Cooper A, et al. FRAX-based
assessment and intervention thresholds—an exploration of thresholds in women aged 50 years
and older in the UK. Osteoporos Int. 2015;26(8):2091–9.
24. Kanis JA, Harvey NC, Cooper C, Johansson H, Oden A, McCloskey EV, et al. A system-
atic review of intervention thresholds based on FRAX : a report prepared for the National
Osteoporosis Guideline Group and the international osteoporosis foundation. Arch Osteoporos.
2016;11(1):25.
25. Lips P. Vitamin D deficiency and secondary hyperparathyroidism in the elderly: consequences
for bone loss and fractures and therapeutic implications. Endocr Rev. 2001;22(4):477–501.
26. MacLaughlin J, Holick MF. Aging decreases the capacity of human skin to produce vitamin
D3. J Clin Invest. 1985;76(4):1536–8.
306 M. Dejaeger et al.
27. Bolland MJ, Grey A, Gamble GD, Reid IR. The effect of vitamin D supplementation on skele-
tal, vascular, or cancer outcomes: a trial sequential meta-analysis. Lancet Diabetes Endocrinol.
2014;2(4):307–20.
28. Jackson RD, LaCroix AZ, Gass M, Wallace RB, Robbins J, Lewis CE, et al. Calcium plus
vitamin D supplementation and the risk of fractures. N Engl J Med. 2006;354(7):669–83.
29. Grant AM, Avenell A, Campbell MK, McDonald AM, MacLennan GS, McPherson GC, et al.
Oral vitamin D3 and calcium for secondary prevention of low-trauma fractures in elderly
people (Randomised Evaluation of Calcium Or vitamin D, RECORD): a randomised placebo-
controlled trial. Lancet. 2005;365(9471):1621–8.
30. Boonen S, Vanderschueren D, Haentjens P, Lips P. Calcium and vitamin D in the prevention
and treatment of osteoporosis - a clinical update. J Intern Med. 2006;259(6):539–52.
31. Bouillon R, Van Schoor NM, Gielen E, Boonen S, Mathieu C, Vanderschueren D, et al.
Optimal vitamin D status: a critical analysis on the basis of evidence-based medicine. J Clin
Endocrinol Metab. 2013;98(8):E1283–304.
32. Eleni A, Panagiotis P. A systematic review and meta-analysis of vitamin D and calcium in
preventing osteoporotic fractures. Clin Rheumatol. 2020;39(12):3571–9.
33. Dawson-Hughes B, Mithal A, Bonjour JP, Boonen S, Burckhardt P, Fuleihan GE, et al.
IOF position statement: vitamin D recommendations for older adults. Osteoporos Int.
2010;21(7):1151–4.
34. Sanders K. ACP journal Club: a single, annual, high dose of oral vitamin D increased falls and
fractures in older women. Ann Intern Med. 2010;153(6):JC3.
35. Bischoff-Ferrari HA, Dawson-Hughes B, Orav EJ, Staehelin HB, Meyer OW, Theiler R, et al.
Monthly high-dose vitamin D treatment for the prevention of functional decline: a randomized
clinical trial. JAMA Intern Med. 2016;176(2):175–83.
36. American Geriatrics Society Workgroup on Vitamin DSfOA. Recommendations abstracted
from the American Geriatrics Society consensus statement on vitamin D for prevention of falls
and their consequences. J Am Geriatr Soc. 2014;62(1):147–52.
37. Rizzoli R, Branco J, Brandi ML, Boonen S, Bruyere O, Cacoub P, et al. Management of osteo-
porosis of the oldest old. Osteoporos Int. 2014;25(11):2507–29.
38. Vondracek SF, Linnebur SA. Diagnosis and management of osteoporosis in the older senior.
Clin Interv Aging. 2009;4:121–36.
39. Black DM, Cummings SR, Karpf DB, Cauley JA, Thompson DE, Nevitt MC, et al. Randomised
trial of effect of alendronate on risk of fracture in women with existing vertebral fractures.
Fracture Intervention Trial Research Group. Lancet. 1996;348(9041):1535–41.
40. Cummings SR, Black DM, Thompson DE, Applegate WB, Barrett-Connor E, Musliner TA,
et al. Effect of alendronate on risk of fracture in women with low bone density but without ver-
tebral fractures: results from the Fracture Intervention Trial. JAMA. 1998;280(24):2077–82.
41. Ensrud KE, Black DM, Palermo L, Bauer DC, Barrett-Connor E, Quandt SA, et al. Treatment
with alendronate prevents fractures in women at highest risk: results from the fracture inter-
vention trial. Arch Intern Med. 1997;157(22):2617–24.
42. Hochberg MC, Thompson DE, Black DM, Quandt SA, Cauley J, Geusens P, et al. Effect of
alendronate on the age-specific incidence of symptomatic osteoporotic fractures. J Bone Miner
Res. 2005;20(6):971–6.
43. Axelsson KF, Wallander M, Johansson H, Lundh D, Lorentzon M. Hip fracture risk and safety
with alendronate treatment in the oldest-old. J Intern Med. 2017;282(6):546–59.
44. Harris ST, Watts NB, Genant HK, McKeever CD, Hangartner T, Keller M, et al. Effects of
risedronate treatment on vertebral and nonvertebral fractures in women with postmenopausal
osteoporosis: a randomized controlled trial. Vertebral Efficacy with Risedronate Therapy
(VERT) Study Group. JAMA. 1999;282(14):1344–52.
45. Reginster J, Minne HW, Sorensen OH, Hooper M, Roux C, Brandi ML, et al. Randomized trial
of the effects of risedronate on vertebral fractures in women with established postmenopausal
osteoporosis. Vertebral Efficacy with Risedronate Therapy (VERT) Study Group. Osteoporos
Int. 2000;11(1):83–91.
21 Pharmacological Treatment of Osteoporosis in Older Patients 307
46. McClung MR, Geusens P, Miller PD, Zippel H, Bensen WG, Roux C, et al. Effect of risedro-
nate on the risk of hip fracture in elderly women. Hip Intervention Program Study Group. N
Engl J Med. 2001;344(5):333–40.
47. Boonen S, McClung MR, Eastell R, El-Hajj Fuleihan G, Barton IP, Delmas P. Safety and
efficacy of risedronate in reducing fracture risk in osteoporotic women aged 80 and older:
implications for the use of antiresorptive agents in the old and oldest old. J Am Geriatr Soc.
2004;52(11):1832–9.
48. Tom SE, Adachi JD, Anderson FA Jr, Boonen S, Chapurlat RD, Compston JE, et al. Frailty
and fracture, disability, and falls: a multiple country study from the global longitudinal study
of osteoporosis in women. J Am Geriatr Soc. 2013;61(3):327–34.
49. Cryer B, Bauer DC. Oral bisphosphonates and upper gastrointestinal tract problems: what is
the evidence? Mayo Clin Proc. 2002;77(10):1031–43.
50. Porras AG, Holland SD, Gertz BJ. Pharmacokinetics of alendronate. Clin Pharmacokinet.
1999;36(5):315–28.
51. Lo JC, Pressman AR, Omar MA, Ettinger B. Persistence with weekly alendronate therapy
among postmenopausal women. Osteoporos Int. 2006;17(6):922–8.
52. Boonen S, Black DM, Colon-Emeric CS, Eastell R, Magaziner JS, Eriksen EF, et al. Efficacy
and safety of a once-yearly intravenous zoledronic acid 5 mg for fracture prevention in
elderly postmenopausal women with osteoporosis aged 75 and older. J Am Geriatr Soc.
2010;58(2):292–9.
53. Black DM, Delmas PD, Eastell R, Reid IR, Boonen S, Cauley JA, et al. Once-yearly zoledronic
acid for treatment of postmenopausal osteoporosis. N Engl J Med. 2007;356(18):1809–22.
54. Lyles KW, Colon-Emeric CS, Magaziner JS, Adachi JD, Pieper CF, Mautalen C, et al.
Zoledronic acid in reducing clinical fracture and mortality after hip fracture. N Engl J Med.
2007;357:nihpa40967.
55. Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, et al. Denosumab
for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med.
2009;361(8):756–65.
56. Boonen S, Adachi JD, Man Z, Cummings SR, Lippuner K, Torring O, et al. Treatment with
denosumab reduces the incidence of new vertebral and hip fractures in postmenopausal women
at high risk. J Clin Endocrinol Metab. 2011;96(6):1727–36.
57. McClung MR, Boonen S, Torring O, Roux C, Rizzoli R, Bone HG, et al. Effect of denosumab
treatment on the risk of fractures in subgroups of women with postmenopausal osteoporosis. J
Bone Miner Res. 2012;27(1):211–8.
58. Papapoulos S, McClung M, Franchimont N, Adachi J, Bone H, Benhamou C, et al. Low frac-
ture incidence is maintained in postmenopausal women ≥75 years with osteoporosis with
long-term denosumab treatment. J Bone Miner Res. 2012;27(Suppl 1)
59. Genant HK, Libanati C, Engelke K, Zanchetta JR, Hoiseth A, Yuen CK, et al. Improvements
in hip trabecular, subcortical, and cortical density and mass in postmenopausal women with
osteoporosis treated with denosumab. Bone. 2013;56(2):482–8.
60. Jamal SA, Ljunggren O, Stehman-Breen C, Cummings SR, McClung MR, Goemaere S, et al.
Effects of denosumab on fracture and bone mineral density by level of kidney function. J Bone
Miner Res. 2011;26(8):1829–35.
61. Tsourdi E, Zillikens MC. Certainties and uncertainties about denosumab discontinuation.
Calcif Tissue Int. 2018;103(1):1–4.
62. Bone HG, Bolognese MA, Yuen CK, Kendler DL, Miller PD, Yang YC, et al. Effects of deno-
sumab treatment and discontinuation on bone mineral density and bone turnover markers in
postmenopausal women with low bone mass. J Clin Endocrinol Metab. 2011;96(4):972–80.
63. Cummings SR, Ferrari S, Eastell R, Gilchrist N, Jensen JB, McClung M, et al. Vertebral
fractures after discontinuation of denosumab: a post hoc analysis of the randomized placebo-
controlled FREEDOM trial and its extension. J Bone Miner Res. 2018;33(2):190–8.
64. Anastasilakis AD, Polyzos SA, Makras P, Aubry-Rozier B, Kaouri S, Lamy O. Clinical fea-
tures of 24 patients with rebound-associated vertebral fractures after denosumab discontinua-
tion: systematic review and additional cases. J Bone Miner Res. 2017;32(6):1291–6.
308 M. Dejaeger et al.
65. Dupont J, Laurent MR, Dedeyne L, Luyten FP, Gielen E, Dejaeger M. Rebound-associated
vertebral fractures after stopping denosumab: report of four cases. Joint Bone Spine.
2020;87(2):171–3.
66. Dupont J, Laurent MR, Dedeyne L, Luyten FP, Gielen E, Dejaeger M. Vertebral fractures after
denosumab cessation. Cleve Clin J Med. 2020;87(6):337–8.
67. Grassi G, Chiodini I, Palmieri S, Cairoli E, Arosio M, Eller-Vainicher C. Bisphosphonates
after denosumab withdrawal reduce the vertebral fractures incidence. Eur J Endocrinol.
2021;185(3):387–96.
68. Solling AS, Harslof T, Langdahl B. Treatment with zoledronate subsequent to denosumab in
osteoporosis: a 2-year randomized study. J Bone Miner Res. 2021;36(7):1245–54.
69. Low A. Should all elderly women receive bisphosphonates to prevent osteoporotic fractures?
Can J Hosp Pharm. 2012;65(1):45–8.
70. Sanchez-Rodriguez D, Bergmann P, Body JJ, Cavalier E, Gielen E, Goemaere S, et al. The
Belgian Bone Club 2020 guidelines for the management of osteoporosis in postmenopausal
women. Maturitas. 2020;139:69–89.
71. Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Reginster JY, et al. Effect of para-
thyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with
osteoporosis. N Engl J Med. 2001;344(19):1434–41.
72. Boonen S, Marin F, Mellstrom D, Xie L, Desaiah D, Krege JH, et al. Safety and efficacy of
teriparatide in elderly women with established osteoporosis: bone anabolic therapy from a
geriatric perspective. J Am Geriatr Soc. 2006;54(5):782–9.
73. Miller PD, Hattersley G, Riis BJ, Williams GC, Lau E, Russo LA, et al. Effect of abaloparatide
vs placebo on new vertebral fractures in postmenopausal women with osteoporosis: a random-
ized clinical trial. JAMA. 2016;316(7):722–33.
74. McClung MR, Harvey NC, Fitzpatrick LA, Miller PD, Hattersley G, Wang Y, et al. Effects of
abaloparatide on bone mineral density and risk of fracture in postmenopausal women aged 80
years or older with osteoporosis. Menopause. 2018;25(7):767–71.
75. Cosman F, Hattersley G, Hu MY, Williams GC, Fitzpatrick LA, Black DM. Effects of
Abaloparatide-SC on fractures and bone mineral density in subgroups of postmeno-
pausal women with osteoporosis and varying baseline risk factors. J Bone Miner Res.
2017;32(1):17–23.
76. Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S, et al. Romosozumab
treatment in postmenopausal women with osteoporosis. N Engl J Med. 2016;375(16):1532–43.
77. Lewiecki EM, Dinavahi RV, Lazaretti-Castro M, Ebeling PR, Adachi JD, Miyauchi A, et al.
One year of romosozumab followed by two years of denosumab maintains fracture risk reduc-
tions: results of the FRAME extension study. J Bone Miner Res. 2019;34(3):419–28.
78. Saag KG, Petersen J, Brandi ML, Karaplis AC, Lorentzon M, Thomas T, et al. Romosozumab
or alendronate for fracture prevention in women with osteoporosis. N Engl J Med.
2017;377(15):1417–27.
79. Watts NB, Adler RA, Bilezikian JP, Drake MT, Eastell R, Orwoll ES, et al. Osteoporosis
in men: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab.
2012;97(6):1802–22.
80. Nayak S, Greenspan SL. Osteoporosis treatment efficacy for men: a systematic review and
meta-analysis. J Am Geriatr Soc. 2017;65(3):490–5.
81. Boonen S, Reginster JY, Kaufman JM, Lippuner K, Zanchetta J, Langdahl B, Rizzoli R,
Lipschitz S, Dimai HP, Witvrouw R, Eriksen E, Brixen K, Russo L, Claessens F, Papanastasiou
P, Antunez O, Su G, Bucci-Rechtweg C, Hruska J, Incera E, Vanderschueren D, Orwoll
E. N. Fracture risk and zoledronic acid therapy in men with osteoporosis. Engl J Med.
2012;367(18):1714–23.
82. Orwoll E, Teglbjaerg CS, Langdahl BL, Chapurlat R, Czerwinski E, Kendler DL, et al. A ran-
domized, placebo-controlled study of the effects of denosumab for the treatment of men with
low bone mineral density. J Clin Endocrinol Metab. 2012;97(9):3161–9.
21 Pharmacological Treatment of Osteoporosis in Older Patients 309
83. Orwoll ES, Scheele WH, Paul S, Adami S, Syversen U, Diez-Perez A, et al. The effect of
teriparatide [human parathyroid hormone (1-34)] therapy on bone density in men with osteo-
porosis. J Bone Miner Res. 2003;18(1):9–17.
84. Lewiecki EM, Blicharski T, Goemaere S, Lippuner K, Meisner PD, Miller PD, et al. A phase
III randomized placebo-controlled trial to evaluate efficacy and safety of romosozumab in men
with osteoporosis. J Clin Endocrinol Metab. 2018;103(9):3183–93.
85. Strom O, Lauppe R, Ljunggren O, Spangeus A, Ortsater G, O'Kelly J, et al. Real-world effec-
tiveness of osteoporosis treatment in the oldest old. Osteoporos Int. 2020;31(8):1525–33.
86. Deardorff WJ, Cenzer I, Nguyen B, Lee SJ. Time to benefit of bisphosphonate therapy for the
prevention of fractures among postmenopausal women with osteoporosis: a meta-analysis of
randomized clinical trials. JAMA Intern Med. 2022;182(1):33–41.
87. Gielen E, Bergmann P, Bruyere O, Cavalier E, Delanaye P, Goemaere S, et al. Osteoporosis in frail
patients: a consensus paper of the Belgian Bone Club. Calcif Tissue Int. 2017;101(2):111–31.
88. Eurostat; 2017. https://ec.europa.eu/eurostat/web/products-eurostat-news/-/EDN-20170930-1
?inheritRedirect=true&redirect=%2Feurostat%2F.
Chronic Respiratory Disease: COPD, IPF
22
Raffaele Antonelli Incalzi and Filippo Luca Fimognari
22.1 Introduction
R. A. Incalzi (*)
Department of Internal Medicine and Geriatrics, Campus Bio-Medico University, Rome, Italy
e-mail: [email protected]; [email protected]
F. L. Fimognari
Department of Medicine and Unit of Geriatrics, Azienda Ospedaliera “Annunziata, Mariano
Santo, S. Barbara”, Cosenza, Italy
Table 22.2 Main problems affecting the diagnosis of exacerbation or limiting the efficacy of
therapy of COPD in the elderly
Coexistent OSASs Frequently it manifests with nocturia due to brisk changes in abdominal
pressure and, then, is misinterpreted as a prostatic symptom
Cognitive Attention should be paid to tailor the therapeutic plan to the individual
impairment/linguistic capabilities and to provide a follow-up
barriers
Impaired dexterity As above
Coexisting CHF Nt-proBNP should be measured and, if high, cardiological work-up
performed
Malnutrition MNA might be used as a screening test
Anorexia Pharyngeal or esophageal mycosis is a frequently missed cause of
anorexia with or without dysphagia
Atypical exacerbation Delirium, gait problems, lethargy, CHF-like presentation, no or mild
dyspnea, thoracic pain may confound the examiner
Arrhythmias Atrial fibrillation, flutter, atrial chaotic tachycardia could account for
hemodynamic instability and require urgent dedicated intervention
besides the correction of the blood gas derangement
Concealed chronic An important cause of fluid retention and a risk factor for drug toxicity
kidney disease
Frequently defective Mainly in patients with diabetes or hyposmia it could make the patient
enteroception unaware of her/his true health status
22.3.1 Education
Making the patient aware of her/his disease, able to recognize significant changes
and to disregard minor ones, and able to adopt pertinent behaviors are a primary
therapeutic objective. Indeed, empowerment and promotion of the enteroception
have been associated with decreased frequency and more timely treatment of exac-
erbations as well as with better disease-related health status [5]. Furthermore, edu-
cation decreases the risk of adverse drug reactions and the use, mainly the improper
use, of healthcare services. To pursue this objective, either individual education or
group programs proved effective. Furthermore, a well-informed patient is able to
appreciate the individual and cumulative values of each therapeutic measure, which
promotes the adherence to maintenance rehabilitation, periodical control visits, and
so on. Finally, an important objective of education is a periodical check of adher-
ence to and retraining the correct use of inhaler devices. Indeed, it has been demon-
strated that even mild cognitive impairment, as reflected by a MMSE < 24 or a
defective performance in the copy of the two pentagons test, identifies people who
will have difficulty with inhalers [6].
22.3.2 Rehabilitation
age and for any stage of COPD severity [7]. Rehabilitation is effective also during
the acute exacerbations, but it plays a primary role as a continuous aid, “mainte-
nance rehabilitation,” with positive effects on both physical and affective domains
[8]. Also patients with disabling COPD benefit from rehabilitation, else more than
patients with mild to moderate COPD [9]. Besides the classical respiratory rehabili-
tation, worthy of attention are general rehabilitation; upper limb exercise, mainly
for the emphysematous patient; and occupational therapy. If possible, rehabilitation
should be tailored to the individual needs by taking into account the mainly hyper-
secretive or dyspneic trait of COPD, exercise-related fatigue, ability to retain the
inherent information, and mood. Thus, a comprehensive assessment is functional to
plan the best rehabilitation for the individual patient. However, if such an approach
is unavailable, it is well proved that also simple exercise such as walking is effective
at improving exercise tolerance and health status [10]. Finally, selected forms of
rehabilitation, such as rehabilitation of swallowing [11], may be of benefit on indi-
vidual bases.
The mucus changes physical status, from more to less dense, by absorbing kinetic
energy (thixotropism). Thus, exercise itself is the primary promoter of mucus clear-
ance. However, mainly in sarcopenic or disabled hypersecretive patients, some
mechanical aids may improve mucus clearance. Among these is the flutter, which,
by delivering kinetic energy, loosens the mucus from the airway walls and increases
airways patency. A comparable effect is obtained by the oscillating positive expira-
tory pressure devices. Other devices produce a negative pressure in the mouth and,
thus, a favorable gradient for mucus clearance; however, their use should be limited
to patient with an efficient cough reflex. Finally, high-frequency and low-frequency
chest compression devices are simple and totally passive mucus clearance promot-
ers [12]. These tools should not be regarded as an alternative to rehabilitation, but
they have the potential for complementing and speeding it in the hypersecretive
phenotype of COPD, mainly in patients whom early fatigability makes unable to
take the full benefit from rehabilitation.
22.3.4 Vaccinations
Vaccination is a key measure of care for older COPD patients [13]. Annual influenza
vaccination prevents exacerbations and is associated with decreased mortality [14].
Pneumococcal 13-valent polysaccharide vaccine (PPSV13), to be repeated every
5 years, has been proved to prevent acute exacerbations and bacteremia from pneu-
mococcus in the elderly. Current CDC recommendations bend toward PPSV23,
which has been proved to decrease pneumonia in COPD patients under 65 years or
FEV1 <40% or comorbidities. Given that pertussis or whooping cough is a primary
cause and a severity factor of COPD exacerbations, a dTaP/dTPa vaccination is
recommended for patients who were not vaccinated in adolescence. However,
316 R. A. Incalzi and F. L. Fimognari
immunity wanes in a few years and a booster dose should be provided after 10 years
and should be repeated every 10 years. Vaccination anti-herpes zoster is also indi-
cated in people over 65 years. COPD patients are at special risk of immunodepres-
sion to malnutrition, steroid use, and chronic inflammation. The adjuvant zoster
vaccine is safe and effective also in immunocompromised patients. Finally,
COVID-19 vaccine is strongly advocated for COPD patients, given that COPD
qualifies as a severity factor for COVID-19 disease [15].
22.3.5 Sleep Hygiene
22.3.6 Nutrition
Age does not decrease the nutrient needs. Thus, the elderly COPD patient
should receive a balanced and complete diet, not differently from an adult one.
Dietary supplementation shown to improve muscle strength is branched amino
acids and PUFA, though the strength of the evidence is only moderate [23, 24].
In hypercatabolic status characterizing selected patients, a greater caloric and
protein intake should be provided, but several obstacles, like digestive problems
(early satiety, epigastric burn) and sarcopenia (mastication, exhaustion, etc.),
make such an objective difficult to reach. Furthermore, dysphagia is highly
prevalent, though unreported, in severe, mainly emphysematous, COPD, and
can manifest either as malnutrition or as chronic cough and/or ab ingestis
pneumonia.
22.3.7 Pharmacological Therapy
22.3.8 Oxygen Therapy
Oxygen should be prescribed like a drug, i.e., at the correct dosage, not more and not
less. Indeed, excess oxygen may depress the respiratory drive, causing hypercarbia until
carbonarcosis, but in the elderly, it carries an important additional risk, the one of coro-
nary or cerebral vasoconstriction [25]. Thus, efforts should be made to obtain and main-
tain an arterial oxygen saturation (SaO2) of 90–95%; higher values might be associated
with adverse effects. Importantly, percutaneous oxygen saturation (SpO2) is frequently
unreliable in the elderly due to hypotension, fever, tremor, chills, atrial fibrillation, and
other causes. Thus, SpO2 >90% should not deter from performing an arterial blood gas
analysis if one or more of these confounding conditions occur and/or if the inherent
information may be of value to assess the metabolic status, e.g., in patients on high-dose
diuretics or renal failure. Attention should be paid to modulate the inspired fraction of
oxygen according to potentially changing needs with sleep and exercise.
High-flow oxygen therapy is safe and effective in elderly patients who are
severely hypoxemic while on Ventimask, but moderate to severe hypercarbia carries
an important risk of ineffectiveness or respiratory depression. Thus, its effects
should be checked timely and, then, monitored to prevent adverse events. Finally,
for people using oxygen concentrators, it is important to know that the provided
fraction of oxygen varies with the respiratory rate and, for selected models, dramati-
cally declines for increasing respiratory rate [26].
Besides being a vital support in severe acute exacerbation, NIV plays an important role
in the chronic care of elderly COPD patients either as a component of palliative care,
to relieve dyspnea, or as an intermittent aid to patient experiencing fatigue and respira-
tory muscle exhaustion. Thus, it should not be considered a measure dedicated to acute
care; else its use is gaining importance in the chronic patient. There are no age-related
changes in the operating rules; else NIV is highly efficacious in the elderly [27, 28].
However, extra caution is needed to prevent pneumothorax or pneumomediastinum
and CHF worsening due to increased intrathoracic barrage, both of which are more
likely to occur in the elderly. In patients experiencing frequent lung atelectases due to
either hypoexpansion or defective mucus clearance, NIV has the potential for prevent-
ing atelectases. Finally, NIV may be of value to assist the patient proved or likely to
undergo nocturnal depression of the respiratory drive which cannot be compensated by
changing the inspired fraction of oxygen. However, the individual needs should be
carefully scrutinized; in the event of coexisting OSAS, C-PAP should be preferred.
22.3.10 Remote Monitoring
It has been proved to prevent exacerbations and improve the quality of life, but only
few studies reach high-quality standards. Nevertheless, there is evidence that a
320 R. A. Incalzi and F. L. Fimognari
22.3.11 Palliative Care
Dyspnea and, to a lesser extent, pain are the two symptoms which characterize end
stage COPD. Being deeply generated and phylogenetically ancient sensations, they
are perceived also in end-stage COPD. Thirst and hunger also are “hard” sensation.
Thus, the end-stage COPD patients should be the object of a comprehensive care
aimed at relieving these untoward sensations through a properly tailored strategy.
However, the principles and practice of palliative care should not be the patrimony
of preterminal care. It has been proved that even in patients with severe COPD,
mainly of the emphysematous type, but not in preterminal stage, using these prin-
ciples and practice can significantly improve the health status. For instance, low-
dose morphine has the potential for relieving dyspnea without causing major
respiratory depression. Furthermore, it can improve the amount and quality
of sleep.
GOLD recommendations apply to patients of any age. However, selected points are
worthy of comment for elderly COPD patients:
9. Hypoxia, malnutrition, and immobility make the older patient at special risk of
developing pressure ulcers. Thus, preventative measures are highly
recommended.
10. A severely hypoxic and normo- or hypocarbic exacerbation should raise the
suspicion of pulmonary embolism or heart failure.
11. Once a worsening of the lung problem and a cardiac complication has been
excluded, a sudden increase or worsening of hypercarbia likely reflects muscle
exhaustion.
12. In older COPD patients hospitalized for severe exacerbations, any effort should
be made to guarantee family and social contacts and to provide a supportive
environment as a strategy preventing delirium.
The interstitial lung diseases (ILDs) constitute a group of parenchymal lung dis-
eases that affect the interstitial space with different grades of inflammation and
fibrosis (Table 22.4). Some ILDs are secondary to known etiologies, including drug
acute exacerbation of IPF, and other conditions. When OP occurs without a spe-
cific cause, COP is diagnosed. There is no association with smoking, and patients
have a mean age of 55 years. The HRTC pattern is represented by peripheral
bilateral (but also unilateral) and diffuse ground-glass opacities or alveolar con-
solidations with air bronchograms, which are often migratory, spare the subpleu-
ral space, more frequently involve the lower lobes, and can spontaneously
disappear. COP is known for its excellent response to corticosteroids, although
relapse is common with corticosteroid tapering or discontinuation [35]. AIP is
similar to the acute respiratory distress syndrome (ARDS), but while ARDS is
provoked by identifiable associated conditions, AIP is idiopathic. Mean age of
patients is 50 years, and there is no relation with smoking. AIP develops as a
severe form of diffuse alveolar damage (DAD), and mortality is around 50%.
Management relies on supportive care, even though steroid therapy is often tried,
without any demonstrated benefit. HRTC pattern is characterized by bilateral,
basal, ground-glass attenuation or consolidation in the dependent areas, in the late
phase followed by architectural distortion, traction bronchiectasis, and honey-
combing in the nondependent areas [31, 32]. Very rare idiopathic ILDs are lym-
phoid interstitial pneumonia (LIP: ground-glass opacities at HRTC and polyclonal
lymphocytes in BAL, good response to corticosteroids) and idiopathic pleuropa-
renchymal fibroelastosis (PPFE, death in 40% of patients). A category of unclas-
sifiable idiopathic interstitial pneumonias [31, 32] completes the idiopathic ILDs
classification (Table 22.4).
Idiopathic ILDs are rare, but their histopathologic and HRTC patterns (UIP,
NSIP, OP, LIP, DAD, and others) represent “prototypes” recurring in more frequent
ILDs, including those associated with CTDs, drugs, vasculitis, and in the hypersen-
sitivity pneumonitis (HP). NSIP is common in most CTDs, particularly SSc, but in
RA and vasculitis, the UIP pattern predominates [36]. HP is an immune-mediated
inflammatory and fibrotic pulmonary response to the inhalation of several antigens
(mainly organic, including avian and microbial agents). HP has been recently cate-
gorized as non-fibrotic HP (ground-glass opacities and mosaic attenuation, associ-
ated with centrilobular nodules on inspiratory images or air trapping in expiratory
images) and fibrotic HP. The fibrotic phenotype is clinically more severe, and
patients are often older [37].
22.5.2 Therapeutic Approach
of forced vital capacity (FVC) in IPF patients by about 50%, without affecting the
time to the first IPF acute exacerbation [39–41]. The mean age of patients enrolled
in the INPULSIS 1 and INPULSIS 2 trials was around 67 years, and eligibility
criteria included age ≥40 years, FVC ≥50%, and diffusing capacity of the lung for
carbon monoxide (DLCO) ≥30% [39]. An analysis of prespecified subgroups from
the INPULSIS trials demonstrated a similar benefit of nintedanib in older
(≥65 years) compared to younger (<65 years) patients [42]. Diarrhea was the
most common adverse effect but caused drug discontinuation in less than 5% of
study patients [39]. Nintedanib is a substrate of P-glycoprotein (P-gp) and
CYP3A4, and concurrent use of P-gp and CYP3A4 inhibitors (e.g., ketoconazole)
or inducers (e.g., carbamazepine) may, respectively, reduce or increase nintedanib
exposure [43].
Pirfenidone, an orally administered pyridine, regulates the expression of TGF-β
and inhibits collagen synthesis, exerting anti-inflammatory, antioxidant, and anti-
fibrotic actions. The recommended dosage of 801 mg 3 times per day is reached
after a 2-week dose titration starting from 267 mg 3 times daily on the first day [43].
In the ASCEND trial, pirfenidone treatment was associated—compared to pla-
cebo—with a reduced rate of FVC decline (by about 45%), a lower proportion of
patients who reported a decreased distance at the 6-min walk test, and with an
increased progression-free survival (neither disease progression nor death) [44].
Pooled data from ASCEND and CAPACITY [45] trials demonstrated lower overall
and IPF-related mortality in the pirfenidone group [44]. The two trial-enrolled
patients aged between 40 and 80 years (mean age was about 67 years), who had
baseline FVC ≥50%, together with DLCO ≥30% in ASCEND and ≥35% in
CAPACITY. However, a recent post hoc analysis from ASCEND and CAPACITY
trials that included a small sample of patients who had more impaired respiratory
function (FVC <50% and/or DLCO <35%) and a mean age of 70 years confirmed
pirfenidone-related clinical benefits, including lower FVC decline and decreased
all-cause mortality [46]. Possible adverse effects are gastrointestinal symptoms
(nausea, dyspepsia), photosensitivity rash, and elevation in aminotransferase level;
these side effects led to discontinuation of study drug in less than 3% of patients
enrolled in the ASCEND trial. Pirfenidone is metabolized by CYP1A2, and the
concurrent use of CYP1A2 inhibitors (e.g., fluvoxamine, ciprofloxacin) should war-
rant dosage reduction [43].
Nintedanib and pirfenidone provide similar benefits in terms of reduction of
FVC decline: the choice of administering one agent or the other should be individu-
alized according to patient’s preference, age, and possible side effects. Since the
eligibility criteria of clinical trials (FVC >50%; DLCO >30–35%; age less than
80 years for pirfenidone) were translated into clinical practice as clinical conditions
required for authorizing treatment, these agents are prescribed only to patients with
mild to moderate IPF, and very elderly patients with advanced disease, comorbidi-
ties, and respiratory failure are always excluded from such treatments. In addition,
possible drug-drug interactions are an obvious concern in frail patients affected by
several illnesses and treated with multiple drugs [30].
22 Chronic Respiratory Disease: COPD, IPF 325
comorbidities and not hastily attributed to progressive lung fibrosis. IPF is prevalent
in the elderly, but also CTD-ILDs and occupational or environmental ILDs tend to
manifest clinically with advancing age. Differential diagnosis should be accurate,
because the standard treatment of IPF (nintedanib or pirfenidone) is different from
that of non-IPF ILDs (immunosuppression). HRTC is a crucial diagnostic tool, but
the current gold standard for ILD diagnosis is a multidisciplinary discussion [33].
Once a diagnosis of non-IPF ILD is obtained in elderly patients, the expected ben-
efits of corticosteroids and/or immunosuppressive agents should be weighed against
possible drug-related complications, comorbidities, overall prognosis estimated by
the comprehensive geriatric assessment, and patient’s expectations. If a confident
diagnosis of IPF is ascertained, nintedanib or pirfenidone should not be denied to
eligible older patients with mild to moderate IPF, accompanied by a strict monitor-
ing of possible drug-related side effects and drug-drug interactions. The gastro-
esophageal reflux disease is highly prevalent in IPF, and episodes of reflux with
microaspiration are believed to contribute to IPF pathogenesis and to the onset of
acute exacerbations. Since a survival benefit was suggested in patients treated with
antiacid agents [59], this treatment should be considered in all IPF patients.
Pulmonary rehabilitation is a crucial intervention in the symptom-centered manage-
ment of IPF and improves the capacity of exercise [60]. High-flow nasal cannula
oxygen therapy is gaining a role as end-of-life palliative treatment of patients requir-
ing very high flow of oxygen that cannot be guaranteed by conventional oxygen
therapy [61] and may facilitate the transition from hospital to home in frail end-
stage ILD patients.
References
1. Kumar M, et al. Senescence-associated secretory phenotype and its possible role in chronic
obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2014;51:323–33.
2. Melloni B, Mounier L, Laaban JP, Chambellan A, Foret D. Muir JF home-based care evolu-
tion in chronic respiratory failure between 2001 and 2015 (Antadir federation observatory).
Respiration. 2018;96:446–54.
3. Bellia V, et al. Quality control of spirometry in the elderly. The SA.R.A. study. Salute
Respiration nell’Anziano = Respiratory Health in the Elderly. Am J Respir Crit Care Med.
2000;161:1094–100.
4. Paladini L, et al. Procalcitonin vs clinical and chest film findings to diagnose community-
acquired pneumonia in patients with acute asthma or acute exacerbations of chronic bronchitis.
Chest. 2011;140:1667–8.
5. Effing T, et al. 2007 Self-management education for patients with chronic obstructive pulmo-
nary disease. Cochrane Rev. 2007:CD002990.
6. Board M, et al. A simple drawing test to identify patients who are unlikely to be able to learn
to use an inhaler. Int J Clin Pract. 2006;60:510.
7. Corhay JL, et al. Should we exclude elderly patients with chronic obstructive pulmonary
disease from a long-time ambulatory pulmonary rehabilitation programme? J Rehabil Med.
2012;44:466–72.
8. Alexopoulos GS, et al. Outcomes of depressed patients undergoing inpatient pulmonary reha-
bilitation. Am J Geriatr Psych. 2006;5:466–75.
9. Di Meo F, et al. Age does not hamper the response to pulmonary rehabilitation of COPD
patients. Age Ageing. 2008;37:530–5.
328 R. A. Incalzi and F. L. Fimognari
10. Maltals F, et al. Effects of home-based pulmonary rehabilitation in patients with chronic
obstructive pulmonary disease: a randomized trial. Ann Intern Med. 2008;149:869–78.
11. Diez Gross R, et al. The coordination of breathing and swallowing in chronic obstructive pul-
monary disease. Am J Respir Crit Care Med. 2009;179:559.
12. O’Clock G, et al. High-frequency and low-frequency chest compression: effects on lung water
secretion, mucus transport, heart rate, and blood pressure using a trapezoidal source pressure
waveform. IEEE Trans Biomed Eng. 2012;59:106–14.
13. Antonelli Incalzi R, et al. Manifesto on the value of adult immunization: “we know, we intend,
we advocate”. Vaccines (Basel). 2021;9(11):1232.
14. Ontario Health Technology Assessment Series; Vol. 12(3), pp. 1–64; 2012.
15. Higham A, Mathioudakis A, Vestbo J, Singh D. COVID-19 and COPD: a narrative review of
the basic science and clinical outcomes. Eur Respir Rev. 2020;29(158):200199.
16. Bellia V, Catalano F, Scichilone N, Incalzi RA, Spatafora M, Vergani C, Rengo F. Sleep dis-
orders in the elderly with and without chronic airflow obstruction: the SARA study. Sleep.
2003;26:318–23.
17. Endeshaw YW, et al. Sleep-disordered breathing and nocturia in older adults.
JAGS. 2004;52:957–60.
18. Wichniak A, Wierzbicka A, Walęcka M, Jernajczyk W. Effects of antidepressants on sleep.
Curr Psychiatry Rep. 2017;19:63.
19. Antonelli Incalzi R et al. Nutritional status in aging and lung disease. In Molecular basis of
nutrition and aging: a volume in the molecular nutrition series; 2016, pp. 411–421.
20. Bhasin S, et al. Sarcopenia definition: the position statements of the sarcopenia definition and
outcomes consortium. JAGS. 2020;68:1410–8.
21. Cruz-Jentoft AJ, Sayer AA. Sarcopenia. Lancet. 2019;393:2636–46.
22. Perkisas S, et al. Application of ultrasound for muscle assessment in sarcopenia: 2020
SARCUS update. Eur Geriatr Med. 2021;12:45–59.
23. Engelen MP, Rutten EP, De Castro CL, Wouters EF, Schols AM, Deutz NE. Supplementation
of soy protein with branched-chain amino acids alters protein metabolism in healthy elderly
and even more in patients with chronic obstructive pulmonary disease. Am J Clin Nutr.
2007;85:431–9.
24. Broekhuizen R, Wouters EF, Creutzberg EC, Weling-Scheepers CA, Schols
AM. Polyunsaturated fatty acids improve exercise capacity in chronic obstructive pulmonary
disease. Thorax. 2005;60:376–82.
25. Moradkhan R, et al. Revisiting the role of oxygen therapy in cardiac patients.
JACC. 2010;56:1013–6.
26. Dunne PJ. The clinical impact of new long-term oxygen therapy technology. Respir Care.
2009;54:1100–11.
27. Comer DM, et al. Domiciliary non-invasive ventilation in the elderly. Effective, tolerated and
justified. Ulster Med J. 2015;84:22–5.
28. Laudisio A, et al. Noninvasive ventilation in older adults admitted to a pneumogeriatric unit.
JAGS. 2014;62:1195.
29. Pedone C, et al. Efficacy of multiparametric telemonitoring on respiratory outcomes in elderly
people with COPD: a randomized controlled trial. BMC: Health Services Res. 2013;13:82.
30. Jo HE, Randhawa S, Corte TJ, Moodley Y. Idiopathic pulmonary fibrosis and the elderly: diag-
nosis and management considerations. Drugs Aging. 2016;33:321–34.
31. American Thoracic Society; European Respiratory Society. American Thoracic Society/
European Respiratory Society international multidisciplinary consensus classification of the
idiopathic interstitial pneumonias. Am J Respir Crit Care Med. 2002;165:277–304.
32. Travis WD, Costabel U, Hansell DM, King TE Jr, Lynch DA, Nicholson AG, et al. ATS/ERS
Committee on idiopathic interstitial pneumonias. An official American Thoracic Society/
European Respiratory Society statement: update of the international multidisciplinary classifi-
cation of the idiopathic interstitial pneumonias. Am J Respir Crit Care Med. 2013;188:733–48.
33. Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, et al. American
Thoracic Society, European Respiratory Society, Japanese Respiratory Society, and Latin
22 Chronic Respiratory Disease: COPD, IPF 329
53. Flaherty KR, Wells AU, Cottin V, Devaraj A, Walsh SLF, Inoue Y, et al. INBUILD trial
investigators. Nintedanib in progressive fibrosing interstitial lung diseases. N Engl J Med.
2019;381:1718–27.
54. Distler O, Highland KB, Gahlemann M, Azuma A, Fischer A, Mayes MD, et al. SENSCIS trial
investigators. Nintedanib for systemic sclerosis-associated interstitial lung disease. N Engl J
Med. 2019;380:2518–28.
55. Maher TM, Corte TJ, Fischer A, Kreuter M, Lederer DJ, Molina-Molina M, et al. Pirfenidone
in patients with unclassifiable progressive fibrosing interstitial lung disease: a double-blind,
randomised, placebo-controlled, phase 2 trial. Lancet Respir Med. 2020;8:147–57.
56. Behr J, Prasse A, Kreuter M, Johow J, Rabe KF, Bonella F, et al. RELIEF investigators.
Pirfenidone in patients with progressive fibrotic interstitial lung diseases other than idiopathic
pulmonary fibrosis (RELIEF): a double-blind, randomised, placebo-controlled, phase 2b trial.
Lancet. Respir Med. 2021;9:476–86.
57. Fimognari FL, Pasqualetti P, Moro L, Franco A, Piccirillo G, Pastorelli R, et al. The associa-
tion between metabolic syndrome and restrictive ventilatory dysfunction in older persons. J
Gerontol A Biol Sci Med Sci. 2007;62:760–5.
58. Pedone C, Scarlata S, Chiurco D, Conte ME, Forastiere F, Antonelli-Incalzi R. Association of
reduced total lung capacity with mortality and use of health services. Chest. 2012;141:1025–30.
59. Fidler L, Sitzer N, Shapera S, Shah PS. Treatment of gastroesophageal reflux in patients
with idiopathic pulmonary fibrosis: a systematic review and meta-analysis. Chest.
2018;153:1405–15.
60. Ferreira A, Garvey C, Connors GL, Hilling L, Rigler J, Farrell S, et al. Pulmonary rehabilita-
tion in interstitial lung disease: benefits and predictors of response. Chest. 2009;135:442–7.
61. Fimognari FL, Rizzo M, Cuccurullo O, Cristiano G, Ricchio R, Ricci C, et al. High-flow nasal
cannula oxygen therapy for acute respiratory failure in a non-intensive geriatric setting. Geriatr
Gerontol Int. 2018;18:1652–3.
Diabetes Mellitus
23
Edoardo Mannucci and Daniele Scoccimarro
E. Mannucci (*)
University of Florence, Florence, Italy
Diabetology and Metabolic Diseases, Careggi Hospital, Florence, Italy
e-mail: [email protected]
D. Scoccimarro
Diabetology and Metabolic Diseases, Careggi Hospital, Florence, Italy
Basal-bolus insulin therapy is the only adequate treatment of type 1 diabetes, irre-
spective of age. The management of such treatment in older adults is more problem-
atic: cognitive impairment, visual loss, and reduced ability to perform complex
manual tasks can all increase the risk of inappropriate insulin administration; renal
impairment, hypoglycemia unawareness, and irregularities in food intake increase
the risk of severe hypoglycemia, and frailty further increases the risk of
hypoglycemia-induced falls. Ability to self-manage cannot be readily passed onto
family members, and older adults with type 1 diabetes are increasingly reliant on
care from the community and outpatient structures.
When basal insulin is needed, long-acting analogs (glargine, detemir, and
degludec insulin) are preferable to NPH human insulin, since they guarantee lower
glucose variability and less hypoglycemia [17], thus allowing a more accurate dose
titration [18]. Insulin glargine U-300 and degludec are ultra-long-acting analogs
with a higher half-life and a lower risk for nocturnal hypoglycemia than glargine
U-100, even in people aged ≥75 years [19, 20]. In addition, their longer duration of
action warrants a greater flexibility in the timing of administration, which can be
advantageous for patients needing third-party assistance for drug administra-
tion [21].
For the management of postprandial glycemia, short-acting analogs (lispro,
aspart, glulisine) should be preferred to human regular insulin, since they allow a
better control of early postprandial hyperglycemia with lower risk for late postpran-
dial hypoglycemia [22, 23]. In addition, short-acting analogs can also be adminis-
tered immediately before or even after, meals, instead of 20–30 min in advance. In
older patients with irregular food intake, the administration of insulin at the end of
the meal allows for the regulation of insulin dose on the basis of the amount of food
actually eaten.
In patients with type 2 diabetes, insulin should be considered when glycemic
control cannot be achieved otherwise. Insulin therapy schemes should be individu-
alized, based on the patient’s actual needs, as determined by the results of glucose
monitoring. Thus, basal insulin should be prescribed when needed for the control of
fasting glucose and prandial insulin for the management of postprandial glucose
excursions. Once-daily basal insulin facilitates compliance to therapy, particularly
in patients requiring caregiver assistance, and it is therefore often considered the
strategy of choice. On the other hand, the age-related reduction in insulin secretion
334 E. Mannucci and D. Scoccimarro
[24] often requires the use of rapid insulin for postprandial glucose control [25, 26],
which cannot be achieved otherwise. The attempt at reducing the postprandial
increase of blood glucose with of basal insulin would be scarcely effective, and it
would imply a high risk of inter-prandial hypoglycemia.
An adequate education of patients and/or caregivers is an integral part of insulin
therapy, particularly in older adults. Such education should include the technique of
insulin injection, the recognition and treatment of hypoglycemia, and the manage-
ment of insulin therapy in relation to food intake and concurrent acute conditions.
All those skills should be periodically verified [27]. In addition, capillary blood
glucose monitoring—which could be problematic in some elderly patients with
reduction in visual acuity, cognitive dysfunction, or limited motor skills—is needed
in all individuals on insulin therapy.
In older patients, the complexity of the insulin treatment induces to consider
insulin only when an acceptable glucose control cannot be achieved otherwise.
Although insulin is still a precious therapeutic option for many elderly patients, it is
associated with a relevant risk for hypoglycemia, falls, and fractures.
Many drugs other than insulin are currently available for the treatment of type 2
diabetes. Some of those agents provide health benefits (e.g., reduction of cardiovas-
cular risk, nephroprotection, etc.) which outreach the favorable effects of improved
glucose control. However, most of available data on the effects of drugs for type 2
diabetes have been collected in adult non-geriatric populations; the risks and bene-
fits associated with some classes of drugs could be partly different in the elderly.
23.3.1 Biguanides
recommends against the use of metformin with serum creatinine above 1.5 mg/dL
(114.4 mmol/L) in men and above 1.4 mg/dL (106.8 mmol/L) in women, respec-
tively, whereas according to the European Society of Cardiology (ESC) and the
European Association for the Study of Diabetes (EASD), metformin is contraindi-
cated in patient with estimated glomerular filtration ratio (eGFR) below 30 mL/min,
recommending caution and dosage reduction when eGFR is between 30 and 60
mL/min [27, 36]. Notably, creatinine levels do not always adequately reflect renal
function in older patients due to sarcopenia. Moreover, older individuals are at
higher risk for dehydration and acute kidney failure. For these reasons, it is advis-
able not to exceed a daily metformin dose of 1500 mg in patients aged more than
75 years. In addition, patients and caregivers should be instructed to suspend met-
formin in case of prolonged fever, vomiting, and diarrhea.
An additional concern is the development of vitamin B12 deficiency in chronic
metformin users [37, 38], which could contribute to anemia, cognitive dysfunc-
tion [37], and symptomatic peripheral neuropathy [39]. Although there are no
available randomized trials on the topic, some guidelines suggest that vitamin
B12 levels should be monitored yearly in older patients in therapy with metfor-
min [34].
with type 2 diabetes, including those aged ≥70 years [48], but further purposely
designed studies are needed to confirm these data.
GLP-1RAs, with the only exception of oral semaglutide, are administered with
subcutaneous injections, requiring adequate visual, motor, and cognitive skills. On
the other hand, the possibility of once-weekly administration facilitates drug man-
agement by patients and caregivers.
The most frequent side effects of GLP-1RAs are gastrointestinal symptoms, such
as nausea and, less frequently, vomiting, and diarrhea [36]. These effects usually
appear in the first weeks of treatment and decrease over the time with the prosecu-
tion of therapy; an initial dose titration is needed with some molecules of the class
in order to minimize side effects. Data from randomized clinical trials seem to show
similar efficacy and safety across ages, with no significant differences across age
classes, even if there was a trend toward higher rates of discontinuation for adverse
events with increasing age [49, 50].
Since weight loss and reduced appetite are usually associated with GLP1-RAs,
these drugs should not be used in malnourished or cachexic elderly patients [33]. In
addition, even if obesity is associated with frailty [51], weight loss is not always
desirable in the elderly because of an increased risk of sarcopenia, falls, and frac-
tures. For this reason, although GLP1-RAs are a very interesting option for the
treatment of type 2 diabetes, their use in older patients is limited by weight loss;
caution is recommended in nonobese elderly patients.
from observational studies. The theoretical risk of volume depletion suggests cau-
tion in prescribing SGLT2is in very old patients. SGLT2is can be an interesting
option in selected healthy elderly patients with T2DM, particularly if overweight or
with uncontrolled hypertension; on the other hand, these medications are not the
treatment of choice in frail patients, especially when treated with loop diuretics (due
to the increased risk of dehydration and orthostatic hypotension) [52]. Dose adjust-
ments for diuretics and insulin are generally required at the initiation of SGLT2i
treatment [36].
Mycotic genital infection is the most frequent adverse event with SGLT2i.
Treatment with SGLT2 inhibitors has also been associated with a modest increase
in the risk of urinary tract infections [64]. Genitourinary infections are more fre-
quent in women, particularly those with a history of recurrent genital infections
[65], and in current or former smokers [66]. In clinical trials, events were gener-
ally mild to moderate in intensity and rarely lead to discontinuation of treat-
ment [67].
and Parkinson’s disease [83], but to date, there is no available clinical evidence in
humans supporting this hypothesis.
Efficacy, tolerability, and safety of DPP-4i in older patients with impaired kidney
function have been confirmed by randomized controlled trials [71, 72], so that this
class can represent a useful option in patients with declined eGFR.
Side effects are uncommon with DPP-4 inhibitors, which are a very considerable
advantage in the elderly. The possibility of an increased risk of pancreatitis associ-
ated with DPP-4i is controversial [84], but it is advisable not to use these drugs in
patients with a history of pancreatitis. Data from observational and randomized con-
trolled trials also show an increased risk for bullous pemphigoid, in particular for
linagliptin and vildagliptin, but the absolute risk is low [85].
DPP4 inhibitors are probably the most extensively studied class of glucose-
lowering drugs in elderly patients. Available evidences show that they are safe in
older individuals, with low risk of hypoglycemia and neutral effect on body weight.
In addition, they are administered orally once daily, facilitating adherence and man-
agement of therapy. Therefore, despite their limited efficacy on glucose control and
the lack of specific cardiovascular actions, they are an interesting treatment option
in elderly, particularly in non-overweight patients with mainly postprandial hyper-
glycemia, as add-on to metformin, or when metformin is not tolerated or
contraindicated.
23.3.5 Thiazolidinediones
inducing a reduction of bone mass [89]. In postmenopausal women, the use of gli-
tazones is associated with accelerated osteoporosis [90] and increased risk of frac-
tures [91]. This phenomenon is not observed in men, since androgens antagonize
the inhibitory effect of TZDs on osteoblastogenesis [89]. As a consequence, gli-
tazones are not among the drugs of choice in postmenopausal women. Finally, pio-
glitazone has been associated with an increased risk of bladder cancer [92],
prompting a relative contraindication in patients with a familiar or personal history
of bladder cancer.
Pioglitazone should be considered as a second-/third-line therapy in elderly
patients due to its effect on bone and heart failure. It could be an effective option in
selected patients, in particular male patients, provided that ventricular function is
preserved.
Sulfonylureas and glinides promote insulin release by blocking β-cell K-ATP chan-
nels, thus inducing membrane hyperpolarization, which triggers insulin secretion.
Sulfonylureas stimulate insulin secretion irrespective of glucose levels [92], poten-
tially leading to hypoglycemia. In epidemiological studies, the incidence of severe
hypoglycemia with sulfonylureas is not dissimilar from that observed with insulin
in type 2 diabetes [93]. This risk is even greater in elderly patients, in whom the
340 E. Mannucci and D. Scoccimarro
impairment of renal function can alter the pharmacokinetic of many drugs of the
class, and the recognition and early treatment of mild hypoglycemia are often com-
promised. Furthermore, in both randomized clinical trials and observational studies,
sulfonylureas are associated with an increased risk of overall and cardiovascular
mortality [94, 95]. The explanation of this phenomenon is still controversial; it is
possible that the blockade of myocardial and cerebral K-ATP channels similar to
those expressed in β-cells reduces the ability of adapting to ischemia [96, 97]. For
all these reasons, sulfonylureas are excluded by some guidelines [14] and consid-
ered third-line treatment by many others [11–13]. An even greater caution should be
taken in elderly patients, considering the greater risk of hypoglycemia and conse-
quent falls and fractures.
23.4 Conclusion
Individuals aged more than 75 years represent a growing fraction of patients with
type 2 diabetes. Despite the epidemiological relevance of this population, most inter-
vention studies enroll only younger individuals, thus limiting available specific evi-
dence. Clinical reasoning suggests that less ambitious glycemic goals should be
chosen for older patients, in the attempt of limiting undesired side effects of pharma-
cological therapy. In addition, the choice of drugs requires caution, considering exist-
ing comorbidities and other factors (such as social support) possibly affecting the
incidence of drug-related adverse events (Table 23.1). Most guidelines [11, 12, 14]
References
1. Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan
JCN, Mbanya JC, Pavkov ME, Ramachandaran A, Wild SH, James S, Herman WH, Zhang P,
Bommer C, Kuo S, Boyko EJ, Magliano DJ. IDF diabetes atlas: global, regional and country-
level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract.
2022;183:109119.
342 E. Mannucci and D. Scoccimarro
21. Marso SP, et al. Efficacy and safety of Degludec versus glargine in type 2 diabetes. N Engl J
Med. 2017;377(8):723–32.
22. Gómez-Huelgas R, et al. Safety and efficacy of a basal-plus regimen with insulin glargine and
insulin glulisine for elderly patients with high cardiovascular risk and type 2 diabetes mellitus.
Rev Clin Esp. 2017;217(4):201–6.
23. Mannucci E, Monami M, Marchionni N. Short-acting insulin analogues vs. regular human
insulin in type 2 diabetes: a meta-analysis. Diabetes Obes Metab. 2009;11(1):53–9.
24. Helman A, et al. Effects of aging and senescence on pancreatic β-cell function. Diabetes Obes
Metab. 2016;18(Suppl 1):58–62.
25. Giugliano D, Chiodini P, Maiorino MI, Bellastella G, Esposito K. Intensification of insulin
therapy with basal-bolus or premixed insulin regimens in type 2 diabetes: a systematic review
and meta-analysis of randomized controlled trials. Endocrine. 2016;51(3):417–28.
26. Bowering K, et al. Faster Aspart versus insulin Aspart as part of a basal-bolus regimen in
inadequately controlled type 2 diabetes: the onset 2 trial. Diabetes Care. 2017;40(7):951–7.
27. Valencia WM, Florez H. Pharmacological treatment of diabetes in older people. Diabetes Obes
Metab. 2014;16(12):1192–203.
28. Natali A, Ferranini E. Effects of metformin and thiazolidinediones on suppression of hepatic
glucose production and stimulation of glucose uptake in type 2 diabetes: a systematic review.
Diabetologia. 2006;49:434–41.
29. Mulherin AJ, Oh AH, Kim H, Grieco A, Lauffer LM, Brubaker PL. Mechanisms underly-
ing metformin-induced secretion of glucagon-like peptide-1 from the intestinal L cell.
Endocrinology. 2011;152:4610–9.
30. Look AHEAD Research Group. Eight-year weight losses with an intensive lifestyle interven-
tion: the look AHEAD study. Obesity (Silver Spring). 2014;22(1):5–13.
31. Schlender L, Martinez YV, Adeniji C, Reeves D, Faller B, Sommerauer C, Al Qur'an T,
Woodham A, Kunnamo I, Sönnichsen A, Renom-Guiteras A. Efficacy and safety of metfor-
min in the management of type 2 diabetes mellitus in older adults: a systematic review for
the development of recommendations to reduce potentially inappropriate prescribing. BMC
Geriatr. 2017;17(Suppl 1):227.
32. Monami M, Candido R, Pintaudi B, Targher G, Mannucci E. SID-AMD joint panel for Italian
guidelines on treatment of type 2 diabetes. Effect of metformin on all-cause mortality and
major adverse cardiovascular events: an updated meta-analysis of randomized controlled tri-
als. Nutr Metab Cardiovasc Dis. 2021;31(3):699–704.
33. Care D, Suppl SS. 12. Older adults: standards of medical care in diabetes-2020. Diabetes Care.
2020;43(January):S152–62.
34. Leroith D, et al. Treatment of diabetes in older adults: an endocrine society. J Clin Endocrinol
Metab. 2019;104(5)
35. Levy J, Cobas RA, Gomes MB. Assessment of efficacy and tolerability of once-daily extended
release metformin in patients with type 2 diabetes mellitus. Diabetol Metab Syndr. 2010;2:16.
https://doi.org/10.1186/1758-5996-2-16. Erratum in: Diabetol Metab Syndr 2010;2:57.
36. Sesti G, et al. Management of diabetes in older adults. Nutr Metab Cardiovasc Dis.
2018;28(3):206–18.
37. Porter KM, et al. Hyperglycemia and metformin use are associated with B vitamin deficiency
and cognitive dysfunction in older adults. J Clin Endocrinol Metab. 2019;104(10):4837–47.
38. Kancherla V, et al. Long-term metformin therapy and monitoring for vitamin B12 deficiency
among older veterans. J Am Geriatr Soc. 2017;65(5):1061–6.
39. Out M, Kooy A, Lehert P, Schalkwijk CA, Stehouwer CDA. Long-term treatment with metfor-
min in type 2 diabetes and methylmalonic acid: post hoc analysis of a randomized controlled
4.3year trial. J Diabetes Complicat. 2018;32(2):171–8.
40. Drucker DJ. Mechanisms of action and therapeutic application of glucagon-like Peptide-1.
Cell Metab. 2018;27(4):740–56.
41. Meier JJ. GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat
Rev Endocrinol. 2012;8:728–42.
344 E. Mannucci and D. Scoccimarro
42. Anderson J. The pharmacokinetic properties of glucagon-like Peptide-1 receptor agonists and
their mode and mechanism of action in patients with type 2 diabetes. J Fam Pract. 2018;67(6
suppl):S8–S13.
43. Singh S, Wright EE Jr, Kwan AY, Thompson JC, Syed IA, Korol EE, Waser NA, Yu MB,
Juneja R. Glucagon-like peptide-1 receptor agonists compared with basal insulins for the treat-
ment of type 2 diabetes mellitus: a systematic review and meta-analysis. Diabetes Obes Metab.
2017;19(2):228–38.
44. Abd El Aziz MS, Kahle M, Meier JJ, Nauck MA. A meta-analysis comparing clinical effects
of short- or long-acting GLP-1 receptor agonists versus insulin treatment from head-to-head
studies in type 2 diabetic patients. Diabetes Obes Metab. 2017;19(2):216–27.
45. Katsurada K, Yada T. Neural effects of gut- and brain-derived glucagon-like peptide-1 and its
receptor agonist. J Diabetes Investig. 2016;7(Suppl 1):64–9.
46. Kristensen SL, et al. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor ago-
nists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular
outcome trials. Lancet Diabetes Endocrinol. 2019;7(10):776–85.
47. Nreu B, Dicembrini I, Tinti F, Sesti G, Mannucci E, Monami M. Major cardiovascular events,
heart failure, and atrial fibrillation in patients treated with glucagon-like peptide-1 receptor
agonists: an updated meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc
Dis. 2020;30(7):1106–14.
48. Cukierman-Yaffe T, et al. Effect of dulaglutide on cognitive impairment in type 2 diabetes: an
exploratory analysis of the REWIND trial. Lancet Neurol. 2020;19(7):582–90.
49. Frias JP, Bonora E, Nevárez Ruiz L, Hsia SH, Jung H, Raha S, Cox DA, Bethel MA, Konig
M. Efficacy and safety of dulaglutide 3.0 and 4.5 mg in patients aged younger than 65
and 65 years or older: post hoc analysis of the AWARD-11 trial. Diabetes Obes Metab.
2021;23(10):2279–88.
50. Mosenzon O, Miller EM, Warren ML. Oral semaglutide in patients with type 2 diabetes
and cardiovascular disease, renal impairment, or other comorbidities, and in older patients.
Postgrad Med. 2020;132(suppl 2):37–47.
51. Wilson PWF, Kannel WB. Obesity, diabetes, and risk of cardiovascular disease in the elderly.
Am J Geriatr Cardiol. 2002;11(2):119–23, 125.
52. Scheen AJ. Sodium-glucose cotransporter type 2 inhibitors for the treatment of type 2 diabetes
mellitus. Nat Rev Endocrinol. 2020;16(10):556–77.
53. Zaccardi F, et al. Efficacy and safety of sodium-glucose cotransporter 2 inhibitors in type
2 diabetes mellitus: systematic review and network meta-analysis. Diabetes Obes Metab.
2016;18:783–94.
54. Sha S, et al. Effect of the sodium glucose co-transporter 2 inhibitor canagliflozin on plasma
volume in patients with type 2 diabetes mellitus. Diabetes Obes Metab. 2014;16:1087–95.
55. Lambers Heerspink HJ, de Zeeuw D, Wie L, Leslie B, List J. Dapagliflozin a glucose-
regulating drug with diuretic properties in subjects with type 2 diabetes. Diabetes Obes Metab.
2013;15:853–62.
56. Lee PC, Ganguly S, Goh SY. Weight loss associated with sodium-glucose cotransporter-2
inhibition: a review of evidence and underlying mechanisms. Obes Rev. 2018;19:1630–41.
57. Zinman B, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N
Engl J Med. 2015;373(22):2117–28.
58. Zelniker TA, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular
and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular
outcome trials. Lancet. 2019;393(10,166):31–9.
59. Neal B, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J
Med. 2017;377(7):644–57.
60. McMurray JJV, et al. A trial to evaluate the effect of the sodium-glucose co-transporter 2
inhibitor dapagliflozin on morbidity and mortality in patients with heart failure and reduced
left ventricular ejection fraction (DAPA-HF). Eur J Heart Fail. 2019;21(5):665–75.
61. Perkovic V, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl
J Med. 2019;380(24):2295–306.
23 Diabetes Mellitus 345
62. Heerspink HJL, Jongs N, Chertow GM, Langkilde AM, McMurray JJV, Correa-Rotter R,
Rossing P, Sjöström CD, Stefansson BV, Toto RD, Wheeler DC, Greene T. DAPA-CKD Trial
Committees and Investigators. Effect of dapagliflozin on the rate of decline in kidney function
in patients with chronic kidney disease with and without type 2 diabetes: a prespecified analy-
sis from the DAPA-CKD trial. Lancet Diabetes Endocrinol. 2021;9(11):743–54.
63. Cintra R, et al. Inhibition of the sodium-glucose co-transporter 2 in the elderly: clinical and
mechanistic insights into safety and efficacy. Rev Assoc Med Bras. 2019;65:70–86.
64. Wu JH, Foote C, Blomster J, Toyama T, Perkovic V, Sundström J, Neal B. Effects of sodium-
glucose cotransporter-2 inhibitors on cardiovascular events, death, and major safety out-
comes in adults with type 2 diabetes: a systematic review and meta-analysis. Lancet Diabetes
Endocrinol. 2016;4(5):411–9.
65. Tentolouris A, Vlachakis P, Tzeravini E, Eleftheriadou I, Tentolouris N. SGLT2 inhibitors:
a review of their antidiabetic and cardioprotective effects. Int J Environ Res Public Health.
2019;16(16):2965.
66. Scoccimarro D, Cipani G, Dicembrini I, Mannucci E. Cigarette smoking as a predictor of
sodium-glucose cotransporter 2 inhibitor-associated genital infections: a retrospective cohort
study. Diabetes Care. 2021;44(6):e127–8.
67. Kohler S, Zeller C, Iliev H, Kaspers S. Safety and tolerability of empagliflozin in patients with
type 2 diabetes: pooled analysis of phase I-III clinical trials. Adv Ther. 2017;34(7):1707–26.
68. Vella A. Mechanism of action of DPP-4 inhibitors—new insights. J Clin Endocrinol Metab.
2012;97(8):2626–8.
69. Karyekar CS, Ravichandran S, Allen E, Fleming D, Frederich R. Tolerability and efficacy of
glycemic control with saxagliptin in older patients (aged ≥ 65 years) with inadequately con-
trolled type 2 diabetes mellitus. Clin Interv Aging. 2013;8:419–30.
70. Del Prato S, et al. Efficacy and safety of linagliptin in subjects with type 2 diabetes mellitus
and poor glycemic control: pooled analysis of data from three placebo-controlled phase III
trials. J Diabetes Complicat. 2013;27(3):274–9.
71. Barzilai N, et al. Efficacy and tolerability of sitagliptin monotherapy in elderly patients with
type 2 diabetes: a randomized, double-blind, placebo-controlled trial. Curr Med Res Opin.
2011;27(5):1049–58.
72. Barnett AH, Huisman H, Jones R, von Eynatten M, Patel S, Woerle H-J. Linagliptin for patients
aged 70 years or older with type 2 diabetes inadequately controlled with common antidiabetes
treatments: a randomised, double-blind, placebo-controlled trial. Lancet (London, England).
2013;382(9902):1413–23.
73. Zangeneh F, Arora PS, Dyck PJ, Bekris L, Lernmark A, Achenbach SJ, Oberg AL, Rizza
RA. Effects of duration of type 2 diabetes mellitus on insulin secretion. Endocr Pract.
2006;12(4):388–93. https://doi.org/10.4158/EP.12.4.388.
74. Vella A, Bock G, Giesler PD, Burton DB, Serra DB, Saylan ML, Deacon CF, Foley JE, Rizza
RA, Camilleri M. The effect of dipeptidyl peptidase-4 inhibition on gastric volume, satiation
and enteroendocrine secretion in type 2 diabetes: a double-blind, placebo-controlled crossover
study. Clin Endocrinol. 2008;69(5):737–44.
75. Apovian CM, Okemah J, O'Neil PM. Body weight considerations in the management of Type
2 diabetes. Adv Ther. 2019;36(1):44–58.
76. Mannucci E, Nreu B, Montereggi C, Ragghianti B, Gallo M, Giaccari A, Monami M, SID-
AMD Joint Panel for Italian Guidelines on Treatment of Type 2 Diabetes. Cardiovascular
events and all-cause mortality in patients with type 2 diabetes treated with dipeptidyl pep-
tidase-4 inhibitors: an extensive meta-analysis of randomized controlled trials. Nutr Metab
Cardiovasc Dis. 2021;31(10):2745–55.
77. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, Ohman P, Frederich
R, Wiviott SD, Hoffman EB, Cavender MA, Udell JA, Desai NR, Mosenzon O, McGuire
DK, Ray KK, Leiter LA, Raz I, SAVOR-TIMI 53 Steering Committee and Investigators.
Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J
Med. 2013;369(14):1317–26.
346 E. Mannucci and D. Scoccimarro
78. Zannad F, Cannon CP, Cushman WC, Bakris GL, Menon V, Perez AT, Fleck PR, Mehta CR,
Kupfer S, Wilson C, Lam H, White WB, EXAMINE Investigators. Heart failure and mortality
outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a
multicentre, randomised, double-blind trial. Lancet. 2015;385(9982):2067–76.
79. McMurray JJV, Ponikowski P, Bolli GB, Lukashevich V, Kozlovski P, Kothny W, Lewsey JD,
Krum H, VIVIDD Trial Committees and Investigators. Effects of vildagliptin on ventricular
function in patients with type 2 diabetes mellitus and heart failure: a randomized placebo-
controlled trial. JACC Heart Fail. 2018;6(1):8–17.
80. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, Josse R, Kaufman KD,
Koglin J, Korn S, Lachin JM, McGuire DK, Pencina MJ, Standl E, Stein PP, Suryawanshi S,
Van de Werf F, Peterson ED, Holman RR, TECOS Study Group. Effect of sitagliptin on car-
diovascular outcomes in type 2 diabetes. N Engl J Med. 2015;373(3):232–42.
81. Rosenstock J, Perkovic V, Johansen OE, Cooper ME, Kahn SE, Marx N, Alexander JH, Pencina
M, Toto RD, Wanner C, Zinman B, Woerle HJ, Baanstra D, Pfarr E, Schnaidt S, Meinicke T,
George JT, von Eynatten M, McGuire DK, CARMELINA Investigators. Effect of linagliptin
vs placebo on major cardiovascular events in adults with type 2 diabetes and high cardiovas-
cular and renal risk: the CARMELINA randomized clinical trial. JAMA. 2019;321(1):69–79.
82. Rosenstock J, Kahn SE, Johansen OE, Zinman B, Espeland MA, Woerle HJ, Pfarr E, Keller
A, Mattheus M, Baanstra D, Meinicke T, George JT, von Eynatten M, McGuire DK, Marx N,
CAROLINA Investigators. Effect of linagliptin vs glimepiride on major adverse cardiovas-
cular outcomes in patients with type 2 diabetes: the CAROLINA randomized clinical trial.
JAMA. 2019;322(12):1155–66.
83. Paolisso G, Monami M, Marfella R, Rizzo MR, Mannucci E. Dipeptidyl peptidase-4 inhibitors
in the elderly: more benefits or risks? Adv Ther. 2012;29(3):218–33.
84. Dicembrini I, Montereggi C, Nreu B, Mannucci E, Monami M. Pancreatitis and pancreatic
cancer in patients treated with dipeptidyl peptidase-4 inhibitors: an extensive and updated
meta-analysis of randomized controlled trials. Diabetes Res Clin Pract. 2020;159:107981.
85. Silverii GA, Dicembrini I, Nreu B, Montereggi C, Mannucci E, Monami M. Bullous pemphi-
goid and dipeptidyl peptidase-4 inhibitors: a meta-analysis of randomized controlled trials.
Endocrine. 2020;69(3):504–7.
86. Lebovitz HE. Thiazolidinediones: the forgotten diabetes medications. Curr Diab Rep.
2019;19(12):151.
87. Nesto RW, et al. Thiazolidinedione use, fluid retention, and congestive heart failure: a con-
sensus statement from the American Heart Association and American Diabetes Association.
Diabetes Care. 2004;27(1):256–63.
88. Scheen AJ. Combined thiazolidinedione-insulin therapy: should we be concerned about
safety? Drug Saf. 2004;27(12):841–56.
89. Benvenuti S, et al. Rosiglitazone stimulates adipogenesis and decreases osteoblastogenesis in
human mesenchymal stem cells. J Endocrinol Investig. 2007;30(9):RC26–30.
90. McDonough AK, Rosenthal RS, Cao X, Saag KG. The effect of thiazolidinediones on BMD
and osteoporosis. Nat Clin Pract Endocrinol Metab. 2008;4(9):507–13.
91. Kahn SE, et al. Rosiglitazone-associated fractures in type 2 diabetes: an analysis from a diabe-
tes outcome progression trial (ADOPT). Diabetes Care. 2008;31(5):845–51.
92. Lv W, Wang X, Xu Q, Lu W. Mechanisms and characteristics of sulfonylureas and glinides.
Curr Top Med Chem. 2020;20(1):37–56.
93. Mannucci E, Naletto L, Vaccaro G, Silverii A, Dicembrini I, Pintaudi B, Monami M. Efficacy
and safety of glucose-lowering agents in patients with type 2 diabetes: a network meta-
analysis of randomized, active comparator-controlled trials. Nutr Metab Cardiovasc Dis.
2021;31(4):1027–34.
94. Mannucci E, Monami M, Candido R, Pintaudi B, Targher G. SID-AMD joint panel for Italian
Guidelines on Treatment of Type 2 Diabetes. Effect of insulin secretagogues on major cardio-
vascular events and all-cause mortality: a meta-analysis of randomized controlled trials. Nutr
Metab Cardiovasc Dis. 2020;30(10):1601–8.
23 Diabetes Mellitus 347
95. Phung OJ, Schwartzman E, Allen RW, Engel SS, Rajpathak SN. Sulphonylureas and risk of car-
diovascular disease: systematic review and meta-analysis. Diabet Med. 2013;30(10):1160–71.
96. Scognamiglio R, Avogaro A, Vigili de Kreutzenberg S, Negut C, Palisi M, Bagolin E, Tiengo
A. Effects of treatment with sulfonylurea drugs or insulin on ischemia-induced myocardial
dysfunction in type 2 diabetes. Diabetes. 2002;51(3):808–12.
97. Liu R, Wang H, Xu B, Chen W, Turlova E, Dong N, Sun CL, Lu Y, Fu H, Shi R, Barszczyk A,
Yang D, Jin T, Mannucci E, Feng ZP, Sun HS. Cerebrovascular safety of sulfonylureas: the role
of KATP channels in neuroprotection and the risk of stroke in patients with type 2 diabetes.
Diabetes. 2016;65(9):2795–809.
Stroke Therapeutics in the Care of Older
Persons 24
A. Bahk, F. A. Kirkham, Y. T. Ng, and Chakravarthi Rajkumar
Stroke is defined as ‘rapidly developing clinical signs of focal (or global) distur-
bance of cerebral function, with symptoms lasting 24 h or longer or leading to
death, with no apparent cause other than of vascular origin’ [1]. It accounts for
approximately 11% of deaths worldwide [2], and over 116 million years of healthy
life lost are due to stroke-related conditions. In Europe, stroke mortality has declined
in recent years due to improvements in diagnosis and treatment [3], but with chang-
ing demographics and population structure, the projected number of stroke survi-
vors in Europe is expected to increase 27% by 2047 [4].
While 87% of strokes are ischaemic compared to only 13% that are haemor-
rhagic [5], the World Stroke Organisation reports that 51% of all deaths from stroke
occur from haemorrhages [6], defined as intracerebral or subarachnoid haemor-
rhages in the absence of trauma [7]. An estimated 30% of strokes occur in those over
80 years, with this group experiencing a threefold higher 30-day case fatality rate
compared to their younger counterparts [8]. Older adults often present with higher
NIHSS (National Institute of Health Stroke Scale) score which translates to more
severe strokes, increased levels of risk factors [9] and have delayed time to throm-
bolysis administration, when given [10], making this patient group a particularly
important focus when it comes to stroke management.
A. Bahk · F. A. Kirkham · Y. T. Ng
University Hospitals Sussex NHS Foundation Trust, Brighton, East Sussex, UK
e-mail: [email protected]; [email protected]; [email protected]
C. Rajkumar (*)
University Hospitals Sussex NHS Foundation Trust, Brighton, East Sussex, UK
Department of Medicine, Brighton and Sussex Medical School, University of Sussex,
Brighton, UK
e-mail: [email protected]; [email protected]
A number of different classification systems have been proposed. The most com-
monly used system is the TOAST criteria (trial of ORG 10172 in acute stroke treat-
ment), focusing on the likely causative event to divide strokes predominately into
large artery atherosclerotic, cardioembolic or small vessel occlusion [13] while
allowing for undetermined or other causes. Approximately 45% of ischaemic
strokes are due to thrombosis [14] in a large or small vessel, while one fifth are
thought to derive from cardiac emboli [15].
Alternatively, Bamford classification (Table 24.1) relies on clinical findings to
determine the brain territory affected, defining strokes as lacunar (LAC), total (TAC)
or partial anterior (PAC), or posterior (POC) and assigning a letter following CT
imaging (I for infarct, H for haemorrhage or S for syndrome) [16]. The oldest old
are more likely to have large vessel (TAC/PAC) occlusion than small vessel (POC/
LAC) occlusion compared to those under 80 years [17].
24.3 Pathophysiology
The Framingham Stroke Risk Profile (FSRP) is the most commonly used tool for
evaluating stroke risk [23] and has been validated for predicting cerebrovascular
events in older patient groups. It incorporates the following established risk factors:
• Sex
• Age
• Smoking
• Blood pressure (treatment for hypertension or raised SBP)
• Diabetes
• Cardiovascular disease
• Atrial fibrillation
FSRP, hyperlipidaemia, alcohol use, obesity, air pollution and dietary choices
have been shown to contribute to increased risk of stroke [24], while higher lev-
els of physical activity have also been shown to be inversely related to stroke
risk [25].
Additional risk factors have been investigated in recent years. There is some
evidence to suggest that serum biomarkers including advanced glycation end prod-
ucts, CRP and total homocysteine are independently associated with increased risk
of stroke [26]. The role of measurable sex hormones and adipokines has been sug-
gested; however, thus far results are inconclusive. Genetic elements have also been
proposed as tools for identifying those at higher risk of stroke, and the APOEε2
allele has been shown to be protective against ischaemic stroke in those aged
71–79 years, but not in people over 80 years [27]. Psychosocial factors shown to
increase stroke risk include depressive symptoms [28], low psychological well-
being and reduced cognitive function [29]. Finally, low socioeconomic status not
only increases the risk of having a stroke but is also associated with reduced access
to care and poorer outcomes [30].
24.5 Presentation
Stroke in older patients does not always present with typical features, and the term
‘stroke chameleon’ has been used to describe unusual presentations. The oldest
old are more likely to present with falls or reduced mobility and less likely to have
sensory symptoms [31]. Other potential presentations include neuropsychiatric
symptoms (such as confusion, memory loss or reduced GCS), abnormal move-
ments, dizziness or headache [32]. The reasons for such divergent presentations
are manifold, as symptoms may still be evolving with subtly different brain
regions involved, alongside practical difficulties in performing comprehensive
examinations in frail older people and the possibility that dual pathologies may
exist simultaneously in multimorbid adults [33]. Full assessment, however, is
paramount to avoid misdiagnosis or attribution of symptoms to another coinciden-
tal illness (e.g. urinary tract infection or pre-existing mobility issues). The most
common misdiagnoses are delirium, syncope, hypertensive emergencies or sys-
temic infection [34].
24.6 Assessment
The Recognition Of Stroke In the Emergency Room (ROSIER) scale is designed for
use in emergency departments to aid in the diagnosis of stroke [35]. However, as
older patients may present with atypical symptoms, and ‘time is brain’ [36], it is
generally recommended to prioritise brain imaging in the form of CT for all poten-
tial strokes. Table 24.2 outlines the recommended investigations for all stroke
patients being assessed in the acute setting.
24 Stroke Therapeutics in the Care of Older Persons 353
Table 24.2 Investigations for older people presenting with acute stroke
Bedside investigations Blood tests Imaging
History and clinical examination Full blood count CT brain within 1 h of presentation
including NIHSS score
Functional assessment Urea and Chest X-ray
electrolytes
Blood pressure Clotting Consider CT angiography or diffusion-
weighted MRI
12-lead ECG Inflammatory Consider echocardiography and 24-h
markers ambulatory ECG to identify cause
Bedside glucose Lipid profile Consider carotid doppler US
Swallow assessment HbA1c
NIHSS National Institutes of Health Stroke Scale
It has been shown that in older adults, frailty score is a better predictor of stroke
outcome than other measures [37]. Equally, pre-stroke cognitive function is an
important determinant of cognitive recovery [38]. Therefore, it is of particular sig-
nificance to assess pre-morbid frailty and cognition in this patient group.
The most widely used tools for frailty assessment include the Rockwood Clinical
Frailty Scale [39] which relies on the judgement of an experienced clinician; the
Fried Frailty Index [40] which focuses on independently measurable variables such
as BMI and handgrip strength; or the Barthel Index [41] which looks at self-reported
ability to perform activities of daily living.
The modified Rankin Scale (mRS) has been applied within stroke research and
in clinical settings as a measure of disability and dependence after stroke. The scale
ranges from 0 (no symptoms) to 6 (death). It can be used to evaluate pre-stroke dis-
ability with validity in prognostication.
Although it is not possible to measure pre-morbid cognitive function on presen-
tation, it is widely accepted that cognitive assessment should be performed prior to
discharge [42]. Many tools have been validated for use in older adults; however,
there is limited evidence for their use in acute stroke [43]. The Montreal Cognitive
Assessment (MoCA) has been suggested as the most useful for identifying cogni-
tive impairment after stroke [44], offering a validated evaluation that is also feasible
to perform regularly in clinical practice.
The immediate treatment of acute ischaemic strokes targets the occluded cerebral
artery to enable revascularisation and re-establish cerebral blood flow. The use of
thrombolytic agents and mechanical thrombectomy is now widely adopted as stan-
dard practice owing to the overall net benefit in reducing disability and mortality.
Numerous studies have shown thrombolysis treatment with intravenous recom-
binant tissue-plasminogen activator (rt-PA), when delivered within 4.5 h, achieved
significant reduction in disability [46, 47]. This was despite symptomatic intracere-
bral haemorrhage (sICH) of 7.7% and fatal intracerebral haemorrhage of 3.6%
within the first 7 days [48].
The exclusion of patients aged ≥80 from these initial trials has led to uncertainty
of potential benefits in this cohort. A major concern was the theoretically greater
bleeding risk in older patients. The Third International Stroke Trial (IST-3) provided
valuable safety and efficacy data in patients aged ≥80, and subsequent trials indi-
cate that the overall benefit of treatment is comparable to younger patients [49].
While sICH rates are similar across age groups, delay in treatment time, larger
strokes, hyperglycaemia and hypertension is implicated in higher bleeding risk and
considered to be contraindications for thrombolysis. The presence of leukoaraiosis
on brain imaging, which is more commonly seen in older patients, is also linked to
increased risk of thrombolysis-related sICH [50]. However, in general, increasing
age is a predictor of adverse outcome and mortality due to non-haemorrhagic com-
plications, often associated with the ageing process. Multi-morbidity and pre-stroke
frailty are also linked to poor prognosis [51]. Treatment decisions therefore require
careful consideration, balancing the overall benefit and risk on an individual basis.
Research continues in the use of magnetic resonance imaging (MRI) and com-
puted tomography perfusion (CTP) to support thrombolysis in patients with stroke
of unknown time of onset or symptoms beyond 4.5 h [52]. This could potentially
widen the treatment time window.
The efficacy of endovascular therapy in the hyperacute treatment of ischaemic
stroke with anterior proximal large vessel occlusion is well established. Mechanical
devices are deployed to remove the clot and open up the occluded cerebral artery
thereby restoring blood flow (Figs. 24.1 and 24.2). The development of newer gen-
eration mechanical devices and advances in technique has led to lower mortality
rates and a substantial reduction in disability at 90 days [53]. These findings have
also been observed in patients aged ≥80 when compared to those given standard
care [53, 54]. Where possible, it is considered to be the treatment of choice for isch-
aemic strokes with a proximal large vessel occlusion for several reasons. Patients
who would otherwise not be eligible for thrombolysis can be offered treatment
without the bleeding risks associated. These include patients with underlying bleed-
ing tendencies and those on anticoagulation treatment. It can also be performed in
conjunction with thrombolysis to achieve higher rates of recanalisation than throm-
bolysis alone [53]. However, the overall recanalisation rates are lower in older
patients [54] with associated higher rates of adverse outcome and mortality [55].
24 Stroke Therapeutics in the Care of Older Persons 355
Poorer neurological reserve and higher risk of post-stroke complication are sug-
gested to be contributing factors.
Current evidence supports the use of intravenous thrombolysis and mechanical
thrombectomy in the treatment of ischaemic strokes, regardless of age. However,
the utilisation of these treatments remains less in older patients [53, 56]. These deci-
sions are likely to be based on various factors including pre-existing comorbidities
and frailty, perceived risk of complications and the overall benefit. Therefore, deci-
sion to treat should be based on the individual patient to avoid potentially harmful
intervention in patients who are unlikely to achieve a favourable outcome.
Fig. 24.2 Angiography of reperfusion in the right middle cerebral artery after mechanical
thrombectomy
neurocritical care are associated with improved mortality and level of independence
in patients with ICH [45].
Elevated blood pressure in the acute phase is a common physiological response in
ICH. Resultant haematoma enlargement and cerebral oedema are feared complica-
tions and predictors of poor outcome [59]. Initiating intensive blood pressure lowering
has shown to be safe in reducing haematoma size without added risk of neurological
deterioration or recurrent stroke events. While this did not translate to significant
reduction in mortality or major disability at 3 months, improved functional outcome
was observed in ordinal shift analysis of mRS in the INTERACT trials with rapid
lowering of blood pressure to a target systolic of 140 mmHg in the first 6 h [60]. Thus,
other factors are likely to contribute to clinical outcome including time frame from
symptom onset to first brain imaging, the volume of haematoma at presentation and
effects of blood pressure variability in the acute phase [61, 62]. Furthermore, extrapo-
lation of data in the oldest old population should be interpreted with caution in view
of uncertainties in the blood pressure physiology of this age group.
The use of vitamin K antagonists (VKA) such as warfarin at presentation results
in larger haematoma size at baseline with greater risk of haematoma expansion and
24 Stroke Therapeutics in the Care of Older Persons 357
The success of stroke prevention with decline in stroke incidence has been offset by
the rapidly ageing population. The exclusion of older patients in numerous land-
mark clinical trials on stroke prevention has led to a paucity of evidence in the
benefits of secondary prevention measures in this cohort. However, with increasing
life expectancy, even patients aged 80 of average health would benefit from second-
ary stroke prevention [72].
358 A. Bahk et al.
24.9.1 Antiplatelets
24.9.2 Antihypertensives
Hypertension is the single most important modifiable risk factor for both ischaemic
and haemorrhagic stroke, and reduction in blood pressure correlates with lower
stroke risk [77].
In the hyperacute phase, blood pressure can be elevated as normal cerebral auto-
regulation is impaired. Lowering blood pressure may be beneficial in the manage-
ment of ICH; however, in acute ischaemic stroke, elevated blood pressure is
considered to have a neuroprotective effect to improve cerebral perfusion to the
ischaemic areas. Optimum timing to initiate antihypertensive treatment remains
uncertain, and recommendations vary from days to weeks [78]. In TIAs, antihyper-
tensive treatment can be commenced once the diagnosis of hypertension is estab-
lished [79], and there are no specific contraindications.
Comparison data between patients aged ≥65 versus <65 showed comparable risk
reduction [80]. Patients aged ≥80 were underrepresented in this study; however, the
Hypertension in the Very Elderly Trial (HYVET) showed that antihypertensive ther-
apy reduced stroke risk by 30% and all-cause mortality by 21% [81]. Adverse effects
of intensive blood pressure lowering in older patients include cerebral hypoperfusion
and orthostatic hypotension. However, slightly higher blood pressure targets of
150/80 mmHg as seen in the HYVET trial could still achieve satisfactory outcome.
lipoprotein (LDL) cholesterol levels to less than 70 mg/dL (1.8 mmol/L) following
ischaemic stroke or TIA could lower risk of subsequent cardiovascular events [83].
Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, also
known as statins, have proven to have protective benefits in reducing stroke risk by
lowering LDL cholesterol [84]. These benefits have also been reflected in further
studies examining older patients for both primary and secondary prevention and with
high level of tolerability [85]. However, these findings have not been consistently
replicated and only seen to be effective in patients with carotid disease [86].
Furthermore, the use of statins should be guided by the life expectancy of the patient
as its benefits are observed over several years. Newer lipid-lowering agents such as
PCSK9 inhibitors have been utilised in patients who are intolerant to statins. With
higher rates of statin intolerance in the older patient, alternative agents may be con-
sidered, although the evidence of its role in the older population is yet to be evaluated.
Diabetes mellitus is a recognised risk factor for ischaemic stroke and is associated
with a fourfold increase in stroke risk. The prevalence of type 2 diabetes increases
with age, and these patients have a higher propensity for strokes and cardiovascular
disease compared to those with type 1 diabetes [87]. Pre-existing diabetes is associ-
ated with recurrent ischaemic stroke and poorer prognosis [88]. However, intensive
glycaemic control has not been shown to reduce cardiovascular events including
stroke and can lead to adverse outcome [89]. This emphasises the need for an indi-
vidualised approach to glycaemic targets and optimisation of coexisting vascular
risk factors, including hypertension and hyperlipidaemia. The extent of glycaemic
control should be balanced with the risk of major hypoglycaemic events. The cur-
rent recommendation is to aim for a target HbA1c of 48 mmol/mol (6.5%) in type 2
diabetes [90].
24.10 Conclusion
Stroke remains common in older patient groups, and while mortality has declined in
recent years, it is a significant cause of morbidity and impaired function. In older
people, presenting symptoms may be atypical, and time to treatment is often
delayed, potentially resulting in poorer outcomes. With advances in imaging and
novel therapeutic approaches, it is imperative for geriatricians to stay informed of
up-to-date evidence and guidelines on stroke management to maximise opportuni-
ties for intervention both in the acute setting and as secondary prevention.
Management within a hyperacute stroke unit by a multidisciplinary team has been
shown to improve outcomes and encourage holistic care. In the future, targeted
molecular therapies and nanotechnology may enable even greater reductions in
mortality and enable faster detection, more effective reperfusion and prevent the
complications still associated with this important condition.
References
1. Truelsen T. The global burden of cerebrovascular disease: WHO. World Health Organisation.
2006; https://www.who.int/healthinfo/statistics/bod_cerebrovasculardiseasestroke.pdf
2. WHO. The top 10 causes of death 2020. https://www.who.int/news-room/fact-sheets/detail/
the-top-10-causes-of-death.
3. Shah R, Wilkins E, Nichols M, Kelly P, El-Sadi F, Wright FL, et al. Epidemiology report:
trends in sex-specific cerebrovascular disease mortality in Europe based on WHO mortality
data. Eur Heart J. 2019;40(9):755–64.
4. Wafa HA, Wolfe CDA, Emmett E, Roth GA, Johnson CO, Wang Y. Burden of stroke in
Europe: thirty-year projections of incidence, prevalence, deaths, and disability-adjusted life
years. Stroke. 2020;51(8):2418–27.
5. Virani SS, Alonso A, Aparicio HJ, Benjamin EJ, Bittencourt MS, Callaway CW, et al. Heart
disease and stroke Statistics-2021 update: a report from the American Heart Association.
Circulation. 2021;143(8):e254–743.
6. Lindsay MP. Global stroke factsheet 2019. WSO, World Stroke Organisation; 2019.
7. Sacco RL, Kasner SE, Broderick JP, Caplan LR, Connors JJ, Culebras A, et al. An updated
definition of stroke for the 21st century: a statement for healthcare professionals from the
American Heart Association/American Stroke Association. Stroke. 2013;44(7):2064–89.
8. Russo T, Felzani G, Marini C. Stroke in the very old: a systematic review of studies on inci-
dence, outcome, and resource use. J Aging Res. 2011;2011:108785.
9. Navis A, Garcia-Santibanez R, Skliut M. Epidemiology and outcomes of ischemic stroke
and transient ischemic attack in the adult and geriatric population. J Stroke Cerebrovasc Dis.
2019;28(1):84–9.
10. Birnbaum LA, Rodriguez JS, Topel CH, Behrouz R, Misra V, Palacio S, et al. Older stroke
patients with high stroke scores have delayed door-to-needle times. J Stroke Cerebrovasc Dis.
2016;25(11):2668–72.
11. Easton JD, Saver JL, Albers GW, Alberts MJ, Chaturvedi S, Feldmann E, et al. Definition
and evaluation of transient ischemic attack: a scientific statement for healthcare profession-
als from the American Heart Association/American Stroke Association Stroke Council;
Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular Radiology
and Intervention; Council on Cardiovascular Nursing; and the Interdisciplinary Council on
Peripheral Vascular Disease. The American Academy of Neurology affirms the value of this
statement as an educational tool for neurologists. Stroke. 2009;40(6):2276–93.
24 Stroke Therapeutics in the Care of Older Persons 363
37. Evans NR, Wall J, To B, Wallis SJ, Romero-Ortuno R, Warburton EA. Clinical frailty inde-
pendently predicts early mortality after ischaemic stroke. Age Ageing. 2020;49(4):588–91.
38. Smithard DG. Stroke in frail older people. Geriatrics (Basel). 2017;2(3)
39. Wallis SJ, Wall J, Biram RW, Romero-Ortuno R. Association of the clinical frailty scale with
hospital outcomes. QJM. 2015;108(12):943–9.
40. Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, et al. Frailty in older
adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci. 2001;56(3):M146–56.
41. Mahoney FI, Barthel DW. Functional evaluation: the Barthel index. Md State Med
J. 1965;14:61–5.
42. Quinn TJ, Elliott E, Langhorne P. Cognitive and mood assessment tools for use in stroke.
Stroke. 2018;49(2):483–90.
43. McGovern A, Pendlebury ST, Mishra NK, Fan Y, Quinn TJ. Test accuracy of informant-based
cognitive screening tests for diagnosis of dementia and multidomain cognitive impairment in
stroke. Stroke. 2016;47(2):329–35.
44. Burton L, Tyson SF. Screening for cognitive impairment after stroke: a systematic review of
psychometric properties and clinical utility. J Rehabil Med. 2015;47(3):193–203.
45. Langhorne P, Ramachandra S. Organised inpatient (stroke unit) care for stroke: network
meta-analysis. Cochrane Database Syst Rev. 2020;4(4):Cd000197.
46. Wardlaw JM, Murray V, Berge E, del Zoppo GJ. Thrombolysis for acute ischaemic stroke.
Cochrane Database Syst Rev. 2014;2014(7):Cd000213.
47. Emberson J, Lees KR, Lyden P, Blackwell L, Albers G, Bluhmki E, et al. Effect of treatment
delay, age, and stroke severity on the effects of intravenous thrombolysis with alteplase for
acute ischaemic stroke: a meta-analysis of individual patient data from randomised trials.
Lancet. 2014;384(9958):1929–35.
48. Wardlaw JM, Murray V, Berge E, del Zoppo G, Sandercock P, Lindley RL, et al. Recombinant
tissue plasminogen activator for acute ischaemic stroke: an updated systematic review and
meta-analysis. Lancet. 2012;379(9834):2364–72.
49. Sandercock P, Wardlaw JM, Lindley RI, Dennis M, Cohen G, Murray G, et al. The benefits
and harms of intravenous thrombolysis with recombinant tissue plasminogen activator within
6 h of acute ischaemic stroke (the third international stroke trial [IST-3]): a randomised con-
trolled trial. Lancet. 2012;379(9834):2352–63.
50. Kongbunkiat K, Wilson D, Kasemsap N, Tiamkao S, Jichi F, Palumbo V, et al. Leukoaraiosis,
intracerebral hemorrhage, and functional outcome after acute stroke thrombolysis. Neurology.
2017;88(7):638–45.
51. Bhatnagar P, Sinha D, Parker RA, Guyler P, O'Brien A. Intravenous thrombolysis in acute
ischaemic stroke: a systematic review and meta-analysis to aid decision making in patients
over 80 years of age. J Neurol Neurosurg Psychiatry. 2011;82(7):712–7.
52. Thomalla G, Boutitie F, Ma H, Koga M, Ringleb P, Schwamm LH, et al. Intravenous alteplase
for stroke with unknown time of onset guided by advanced imaging: systematic review and
meta-analysis of individual patient data. Lancet. 2020;396(10,262):1574–84.
53. Goyal M, Menon BK, van Zwam WH, Dippel DW, Mitchell PJ, Demchuk AM, et al.
Endovascular thrombectomy after large-vessel ischaemic stroke: a meta-analysis of individ-
ual patient data from five randomised trials. Lancet. 2016;387(10,029):1723–31.
54. Zhao W, Ma P, Zhang P, Yue X. Mechanical thrombectomy for acute ischemic stroke in octo-
genarians: a systematic review and meta-analysis. Front Neurol. 2019;10:1355.
55. Hilditch CA, Nicholson P, Murad MH, Rabinstein A, Schaafsma J, Pikula A, et al.
Endovascular management of acute stroke in the elderly: a systematic review and meta-
analysis. AJNR Am J Neuroradiol. 2018;39(5):887–91.
56. Arora R, Salamon E, Katz JM, Cox M, Saver JL, Bhatt DL, et al. Use and outcomes of
intravenous thrombolysis for acute ischemic stroke in patients ≥90 years of age. Stroke.
2016;47(9):2347–54.
57. Parry-Jones AR, Paley L, Bray BD, Hoffman AM, James M, Cloud GC, et al. Care-limiting
decisions in acute stroke and association with survival: analyses of UK national quality reg-
ister data. Int J Stroke. 2016;11(3):321–31.
24 Stroke Therapeutics in the Care of Older Persons 365
58. Morgenstern LB, Zahuranec DB, Sánchez BN, Becker KJ, Geraghty M, Hughes R, et al. Full
medical support for intracerebral hemorrhage. Neurology. 2015;84(17):1739–44.
59. Rodriguez-Luna D, Piñeiro S, Rubiera M, Ribo M, Coscojuela P, Pagola J, et al. Impact of
blood pressure changes and course on hematoma growth in acute intracerebral hemorrhage.
Eur J Neurol. 2013;20(9):1277–83.
60. Anderson CS, Heeley E, Huang Y, Wang J, Stapf C, Delcourt C, et al. Rapid blood-pressure low-
ering in patients with acute intracerebral hemorrhage. N Engl J Med. 2013;368(25):2355–65.
61. Lattanzi S, Cagnetti C, Provinciali L, Silvestrini M. How should we lower blood pres-
sure after cerebral hemorrhage? A systematic review and meta-analysis. Cerebrovasc Dis.
2017;43(5–6):207–13.
62. Al-Shahi Salman R, Frantzias J, Lee RJ, Lyden PD, Battey TWK, Ayres AM, et al. Absolute
risk and predictors of the growth of acute spontaneous intracerebral haemorrhage: a system-
atic review and meta-analysis of individual patient data. Lancet Neurol. 2018;17(10):885–94.
63. Seiffge DJ, Goeldlin MB, Tatlisumak T, Lyrer P, Fischer U, Engelter ST, et al. Meta-analysis
of haematoma volume, haematoma expansion and mortality in intracerebral haemorrhage
associated with oral anticoagulant use. J Neurol. 2019;266(12):3126–35.
64. Tsivgoulis G, Wilson D, Katsanos AH, Sargento-Freitas J, Marques-Matos C, Azevedo E,
et al. Neuroimaging and clinical outcomes of oral anticoagulant-associated intracerebral
hemorrhage. Ann Neurol. 2018;84(5):694–704.
65. Christensen H, Cordonnier C, Kõrv J, Lal A, Ovesen C, Purrucker JC, et al. European stroke
organisation guideline on reversal of oral anticoagulants in acute intracerebral haemorrhage.
Eur Stroke J. 2019;4(4):294–306.
66. Mendelow AD, Gregson BA, Fernandes HM, Murray GD, Teasdale GM, Hope DT, et al.
Early surgery versus initial conservative treatment in patients with spontaneous supratentorial
intracerebral haematomas in the International Surgical Trial in Intracerebral Haemorrhage
(STICH): a randomised trial. Lancet. 2005;365(9457):387–97.
67. Hanley DF, Thompson RE, Rosenblum M, Yenokyan G, Lane K, McBee N, et al. Efficacy
and safety of minimally invasive surgery with thrombolysis in intracerebral haemorrhage
evacuation (MISTIE III): a randomised, controlled, open-label, blinded endpoint phase 3
trial. Lancet. 2019;393(10175):1021–32.
68. Sondag L, Schreuder F, Boogaarts HD, Rovers MM, Vandertop WP, Dammers R, et al.
Neurosurgical intervention for supratentorial intracerebral Hemorrhage. Ann Neurol.
2020;88(2):239–50.
69. Fonseca AC, Merwick Á, Dennis M, Ferrari J, Ferro JM, Kelly P, et al. European Stroke
Organisation (ESO) guidelines on management of transient ischaemic attack. Eur Stroke
J. 2021;6(2):V
70. Prabhakaran S, Silver AJ, Warrior L, McClenathan B, Lee VH. Misdiagnosis of transient
ischemic attacks in the emergency room. Cerebrovasc Dis. 2008;26(6):630–5.
71. Rothwell PM, Giles MF, Chandratheva A, Marquardt L, Geraghty O, Redgrave JN, et al.
Effect of urgent treatment of transient ischaemic attack and minor stroke on early recurrent
stroke (EXPRESS study): a prospective population-based sequential comparison. Lancet.
2007;370(9596):1432–42.
72. Kaplan RC, Tirschwell DL, Longstreth WT Jr, Manolio TA, Heckbert SR, Lefkowitz D, et al.
Vascular events, mortality, and preventive therapy following ischemic stroke in the elderly.
Neurology. 2005;65(6):835–42.
73. Rothwell PM, Algra A, Amarenco P. Medical treatment in acute and long-term sec-
ondary prevention after transient ischaemic attack and ischaemic stroke. Lancet.
2011;377(9778):1681–92.
74. Rothwell PM, Algra A, Chen Z, Diener HC, Norrving B, Mehta Z. Effects of aspirin on risk
and severity of early recurrent stroke after transient ischaemic attack and ischaemic stroke:
time-course analysis of randomised trials. Lancet. 2016;388(10,042):365–75.
75. CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in
patients at risk of ischaemic events (CAPRIE). Lancet. 1996;348(9038):1329–39.
366 A. Bahk et al.
76. Bhatia K, Jain V, Aggarwal D, Vaduganathan M, Arora S, Hussain Z, et al. Dual antiplatelet
therapy versus aspirin in patients with stroke or transient ischemic attack: meta-analysis of
randomized controlled trials. Stroke. 2021;52(6):e217–e23.
77. Zhang H, Thijs L, Staessen JA. Blood pressure lowering for primary and secondary preven-
tion of stroke. Hypertension. 2006;48(2):187–95.
78. Appleton JP, Sprigg N, Bath PM. Blood pressure management in acute stroke. Stroke Vasc
Neurol. 2016;1(2):72–82.
79. National Institute for Health and Care Excellence (NICE). Stroke and transient ischaemic
attack in over 16s: diagnosis and initial management. NICE guideline [NG128]; 2019.
80. Turnbull F, Neal B, Ninomiya T, Algert C, Arima H, Barzi F, et al. Effects of different regi-
mens to lower blood pressure on major cardiovascular events in older and younger adults:
meta-analysis of randomised trials. BMJ. 2008;336(7653):1121–3.
81. Beckett NS, Peters R, Fletcher AE, Staessen JA, Liu L, Dumitrascu D, et al. Treatment of
hypertension in patients 80 years of age or older. N Engl J Med. 2008;358(18):1887–98.
82. Aronow WS. Managing hyperlipidaemia in the elderly: special considerations for a popula-
tion at high risk. Drugs Aging. 2006;23(3):181–9.
83. Amarenco P, Kim JS, Labreuche J, Charles H, Abtan J, Béjot Y, et al. A comparison of two
LDL cholesterol targets after ischemic stroke. N Engl J Med. 2020;382(1):9.
84. Amarenco P, Labreuche J, Lavallée P, Touboul PJ. Statins in stroke prevention and carotid ath-
erosclerosis: systematic review and up-to-date meta-analysis. Stroke. 2004;35(12):2902–9.
85. Deedwania P, Stone PH, Bairey Merz CN, Cosin-Aguilar J, Koylan N, Luo D, et al. Effects
of intensive versus moderate lipid-lowering therapy on myocardial ischemia in older patients
with coronary heart disease: results of the Study Assessing Goals in the Elderly (SAGE).
Circulation. 2007;115(6):700–7.
86. Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM, Cobbe SM, et al. Pravastatin
in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial.
Lancet. 2002;360(9346):1623–30.
87. Tun NN, Arunagirinathan G, Munshi SK, Pappachan JM. Diabetes mellitus and stroke: a
clinical update. (1948–9358 (Print)).
88. Shou J, Zhou L, Zhu S, Zhang X. Diabetes is an independent risk factor for stroke recurrence
in stroke patients: a meta-analysis. J Stroke Cerebrovasc Dis. 2015;24(9):1961–8.
89. Turnbull FM, Abraira C, Anderson RJ, Byington RP, Chalmers JP, Duckworth WC, et al.
Intensive glucose control and macrovascular outcomes in type 2 diabetes. Diabetologia.
2009;52(11):2288–98.
90. National Institute for Health and Care Excellence (NICE). Type 2 diabetes in adults: manage-
ment. NICE guideline [NG28]; 2015.
91. Friberg L, Rosenqvist M, Lindgren A, Terént A, Norrving B, Asplund K. High prevalence of
atrial fibrillation among patients with ischemic stroke. Stroke. 2014;45(9):2599–605.
92. National Institute for Health and Care Excellence (NICE). Atrial fibrillation: diagnosis and
management. NICE guidelines [NG196]; 2021.
93. Mant J, Hobbs FD, Fletcher K, Roalfe A, Fitzmaurice D, Lip GY, et al. Warfarin versus
aspirin for stroke prevention in an elderly community population with atrial fibrillation (the
Birmingham Atrial Fibrillation Treatment of the Aged Study, BAFTA): a randomised con-
trolled trial. Lancet. 2007;370(9586):493–503.
94. Björck F, Renlund H, Lip GY, Wester P, Svensson PJ, Själander A. Outcomes in a warfarin-
treated population with atrial fibrillation. JAMA Cardiol. 2016;1(2):172–80.
95. Ruff CT, Giugliano RP, Braunwald E, Hoffman EB, Deenadayalu N, Ezekowitz MD, et al.
Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients
with atrial fibrillation: a meta-analysis of randomised trials. Lancet. 2014;383(9921):955–62.
96. Pugh D, Pugh J, Mead GE. Attitudes of physicians regarding anticoagulation for atrial fibril-
lation: a systematic review. Age Ageing. 2011;40(6):675–83.
97. Petty GW, Brown RD Jr, Whisnant JP, Sicks JD, O'Fallon WM, Wiebers DO. Ischemic stroke
subtypes: a population-based study of incidence and risk factors. Stroke. 1999;30(12):2513–6.
24 Stroke Therapeutics in the Care of Older Persons 367
98. MRC European Carotid Surgery Trial: interim results for symptomatic patients with severe
(70-99%) or with mild (0-29%) carotid stenosis. European Carotid Surgery Trialists’
Collaborative Group. Lancet. 1991;337(8752):1235–43.
99. Barnett HJM, Taylor DW, Haynes RB, Sackett DL, Peerless SJ, Ferguson GG, et al. Beneficial
effect of carotid endarterectomy in symptomatic patients with high-grade carotid stenosis. N
Engl J Med. 1991;325(7):445–53.
100. Rothwell PM, Eliasziw M, Gutnikov SA, Fox AJ, Taylor DW, Mayberg MR, et al. Analysis of
pooled data from the randomised controlled trials of endarterectomy for symptomatic carotid
stenosis. Lancet. 2003;361(9352):107–16.
101. Naylor AR, Sillesen H, Schroeder TV. Clinical and imaging features associated with an
increased risk of early and late stroke in patients with symptomatic carotid disease. Eur J
Vasc Endovasc Surg. 2015;49(5):513–23.
102. Bonati LH, Lyrer P, Ederle J, Featherstone R, Brown MM. Percutaneous transluminal bal-
loon angioplasty and stenting for carotid artery stenosis. Cochrane Database Syst Rev.
2012;(9):Cd000515.
103. Bonati LH, Kakkos S, Berkefeld J, de Borst GJ, Bulbulia R, Halliday A, et al. European
Stroke Organisation guideline on endarterectomy and stenting for carotid artery stenosis. Eur
Stroke J. 2021;6(2):I.
104. Langhorne P, Stott DJ, Robertson L, MacDonald J, Jones L, McAlpine C, et al. Medical
complications after stroke: a multicenter study. Stroke. 2000;31(6):1223–9.
105. Cruz-Flores S, Berge E, Whittle IR. Surgical decompression for cerebral oedema in acute
ischaemic stroke. Cochrane Database Syst Rev. 2012;1:Cd003435.
106. Zelano J, Holtkamp M, Agarwal N, Lattanzi S, Trinka E, Brigo F. How to diagnose and treat
post-stroke seizures and epilepsy. Epileptic Disord. 2020;22(3):252–63.
107. Dennis M, Sandercock P, Reid J, Graham C, Forbes J, Murray G. Effectiveness of inter-
mittent pneumatic compression in reduction of risk of deep vein thrombosis in patients
who have had a stroke (CLOTS 3): a multicentre randomised controlled trial. Lancet.
2013;382(9891):516–24.
108. Wu S, Kutlubaev MA, Chun HY, Cowey E, Pollock A, Macleod MR, et al. Interventions for
post-stroke fatigue. Cochrane Database Syst Rev. 2015;2015(7):Cd007030.
109. Pendlebury ST, Rothwell PM. Incidence and prevalence of dementia associated with transient
ischaemic attack and stroke: analysis of the population-based Oxford Vascular Study. Lancet
Neurol. 2019;18(3):248–58.
Optimizing Pharmacotherapy in Older
Patients with Depression or Anxiety 25
Sylvie Bonin-Guillaume
25.1 Depression
Depression occurs up to 11% in the ageing population [1] with numerous atypical
presentations; only 12% of depressed older persons meet the DSM major depressive
criteria [2]. This prevalence reaches 40% in long-term care and nursing homes [3].
Thus, depression in older patient is often underdiagnosed and undertreated,
although it is associated with a poor prognosis (suicide [4], relapse, chronicity [5]).
Depression is also independently associated with frailty [6], malnutrition [7], func-
tional decline [8] and morbidity.
S. Bonin-Guillaume (*)
Internal Medicine and Geriatric Department, Aix Marseille University, University Hospital of
Marseille, Hôpital de Sainte Marguerite, Marseille, France
e-mail: [email protected]; [email protected]
Table 25.1 Recommendations for a better antidepressant treatment efficiency (from [13, 19–21])
1. Discuss the choice of antidepressant with the patient
• First choice: SSRI (selective serotonin reuptake inhibitors)
• Efficiency and tolerability of any antidepressants previously taken
2. Inform the patient about the following:
• Gradual development of the full antidepressant effect
• Importance of taking medication as prescribed and the need to continue beyond remission
• Potential side effects and drug interactions
3. Monitor AD response regularly and side effects carefully
• Do not prescribe sub-therapeutic doses of antidepressants
• Evaluate improvement or remission (a delayed response up to 12 weeks is usual in older
patients)
• Monitor carefully for side effects
4. Check treatment duration and adherence
• >6 months after complete remission for the first episode (optimally 1 year after complete
remission), longer if past history of depression episode
• A step-by-step withdrawal (at least 1 month, optimally 3 months)
5. Avoid psychotropic drugs co-prescription unless:
• Congruent psychosis symptoms present (antipsychotic should be proposed)
• Severe anxiety (benzodiazepines might be associated with 12 weeks maximum)
from clinical trials. However, it has shown to be effective both in treating manic
symptoms and in reducing depressive symptoms.
Although there can be adverse effects with lithium, it is generally well tolerated
in older adults. Regular monitoring of renal function and lithium levels
(0.4–0.8 mmol/L) as well as drug-drug interactions (as diuretics, angiotensin-
converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs) and
non-steroidal anti-inflammatory drugs (NSAIDs) is necessary to prevent side effects
[38]. Some studies have described a positive effect of continuous lithium treatment
in reducing the risk of neurocognitive disorders in older adults [39].
25.2 Anxiety
Benzodiazepines are the first-line treatment for anxiety attacks episodes but are
overused in older populations. Recent studies showed that 60% of older persons on
benzodiazepines are chronic users (over 5 consecutive months) [28], especially
those living in nursing homes [48] or having neurocognitive disorders [29].
Guidelines recommend that benzodiazepines should not be prescribed for more than
12 weeks for acute anxiety that impacts on daily life, and no more than 4 weeks for
sleep disorders [49]. Principles of benzodiazepines prescription are summarized in
Table 25.3.
Table 25.3 Optimizing benzodiazepines prescription in anxiety disorders (from [21, 50])
1. Check the indication (anxiety/insomnia)
2. Inform patients of the treatment effect and treatment duration
3. Inform patient of the drug side effects
4. Prescribe for a maximum of 12 weeks for anxiety including the withdrawal
5. Ensure patient understanding and compliance to the treatment
6. Start with a low dose and keep the lowest dose
7. Choose a short-lasting effect product
8. In case of a long users, propose a step-by-step withdrawal in a specific consultation
376 S. Bonin-Guillaume
It is well known that benzodiazepine has numerous side effects, injurious falls,
fractures, drowsiness, delirium and agitation [50]. Recently, several pharmacologi-
cal studies demonstrated an association between BZD users and the risk of develop-
ing neurocognitive disorders, particularly for chronic users (over 6 months) and for
long-acting BZD users [51]. This risk decreases, but several months after, benzodi-
azepines withdrawal [52].
To avoid benzodiazepine prescription, some non-benzodiazepine drugs may be
used to treat anxiety. They are considered to have fewer side effects because of dif-
ferent pharmacological action. However, no clinical trials have been conducted in old
populations. Buspirone and etifoxine are not commonly used in this population.
Hydroxyzine is more often prescribed for agitation and/or anxiety in older patients.
However, because of a high anticholinergic score and the risk of a QT lengthening
[53], the use of this drug is no more recommended for patients aged 75 years and over.
Benzodiazepine reduction in older patients is effective even in general practice
with low risk of withdrawal symptoms [54]. This should be attempted in chronic
users during a dedicated consultation with a progressive reduction regimen [49].
References
1. Castro Costa E, Dewey M, Stewart R, Barnerjee S, Huppert F, Mendoca-Lima C, et al.
Prevalence of depressive symptoms and syndromes in later life in ten European countries. Br J
Psychiatry. 2007;191:393–401. https://doi.org/10.1192/bjp.bp.107.036772.
2. Judd LL, Akiskal HS. The clinical and public health relevance of current research on sub-
threshold depressive symptoms to elderly patients. Am J Geriatr Psychiatry. 2002;10:233–8.
3. Blazer DG. Epidemiology of depression: prevalence and incidence. In: Copeland JRM, Abou-
Saleh MT, Blazer DG, editors. Principles and practice of geriatric psychiatry. 2nd ed. London:
John Wiley & Sons Ltd; 2002. p. 389–92.
4. Jalenques I, Gallarda T, Vaille-Perret E. Suicide. In: Clément JP, editor. Psychiatrie de la
Personne âgée. Paris: Edition Flammarion; 2010. p. 262–75.
5. Beekman AT, Geerlings SW, Deeg DJ, Smit JH, Schoevers RS, de Beurs E, et al. The natu-
ral history of late-life depression : a 6-year prospective study in the community. Arch Gen
Psychiatry. 2002;59:605–11. https://doi.org/10.1001/archpsyc.59.7.60.
25 Optimizing Pharmacotherapy in Older Patients with Depression or Anxiety 377
6. Vaughan L, Corbin AL, Goveas JS. Depression and frailty in later life: a systematic review.
Clin Interv Aging. 2015;10:1947–58. https://doi.org/10.2147/CIA.S69632.
7. Cabrera MA, Mesas AE, Garcia AR, de Andrade SM. Malnutrition and depression among
community-dwelling elderly people. J Am Med Dir Assoc. 2007;8:582–4. https://doi.
org/10.1016/j.jamda.2007.07.008.
8. Da Silva SA, Scazufca M, Menezes PR. Population impact of depression on functional disabil-
ity inelderly: results from “São Paulo Ageing & Health Study” (SPAH). Eur Arch Psychiatry
Clin Neurosci. 2013;263:153–8. https://doi.org/10.1007/s00406-012-0345-4.
9. Kok RM, Nolen WA, Heeren TJ. Efficacy of treatment in older depressed patients: a systematic
review and meta-analysis of double-blind randomized controlled trials with antidepressants. J
Affect Disord. 2012;141:103–15. https://doi.org/10.1016/j.jad.2012.02.036.
10. Kok RB, Reynolds CF III. Management of depression in older adults: a review.
JAMA. 2017;317:2114–22. https://doi.org/10.1001/jama.2017.5706.
11. Calati R, Salvina Signorelli M, Balestri M, Marsano A, De Ronchi D, Aguglia E, Serretti
A. Antidepressants in elderly: metaregression of double-blind, randomized clinical trials. J
Affect Disord. 2013;147:1–8.
12. Pimontel MA, Rindskopf D, Rutherford BR, Brown PJ, Roose SP, Sneed JR. A meta-analysis
of executive dysfunction and antidepressant treatment response in late-life depression. Am J
Geriatr Psychiatry. 2016;24:31–41.
13. Alamo C, Lopez-Munoz F, Garcia-Garcia P, Garcia-Ramoz S. Risk–benefit analysis of
antidepressant drug treatment in the elderly. Psychogeriatrics. 2014;14:261–8. https://doi.
org/10.1111/psyg.12057.
14. Katona C, Hansen T, Olsen CK. A randomized, doubleblind, placebo-controlled, duloxetine-
referenced, fixed-dose study comparing the efficacy and safety of Lu AA21004 in elderly
patients with major depressive disorder. Int Clin Psychopharmacol. 2012;27:215–23.
15. Bishop MM, Fixen DR, Linnebur SA, Pearson SM. Cognitive effects of vortioxetine in older
adults: a systematic review. Ther Adv Psychopharmacol. 2021;11:20451253211026796.
https://doi.org/10.1177/20451253211026796. eCollection 2021.
16. Zhang X, Cai Y, Hu X, Lu CY, Nie X, Shi L. Systematic review and meta-analysis of vortiox-
etine for the treatment of major depressive disorder in adults. Front Psychiatry. 2022;13:922648.
https://doi.org/10.3389/fpsyt.2022.922648. eCollection 2022.
17. Benraad CE, Kamerman-Celie F, van Munster BC, Oude Voshaar RC, Spijker J, Olde Rikkert
MG. Geriatric characteristics in randomised controlled trials on antidepressant drugs for
older adults: a systematic review. Int J Geriatr Psychiatry. 2016;31:990–1003. https://doi.
org/10.1002/gps.4443.
18. American Psychiatric Association. Diagnostic and statistical manual of mental disorders.
5th ed. Washington DC: American Psychiatric Association; 2013. https://doi.org/10.1016/j.
jagp.2015.05.010.
19. National Institute for health & Care Excellence. Depression in adults: recognition and man-
agement. October 2009, updated April 2016. www.nice.org.uk
20. Haute Autorité de Santé. Recommandations pour la pratique clinique. Episode dépressif car-
actérisé de l’adulte : prise en charge en soins de premier recours (guidelines for the manage-
ment of depressive episode in primary care). October 2017. 45p. www.has.fr. Accessed 02
Nov 2021.
21. Bonin-Guillaume S, Roche J, Vandel P. Psychotropes. In: Clément JP, Calvet B, editors.
Psychiatrie de la personne âgée. 2nd ed. Paris: Lavoisier; 2019. p. 423–42.
22. Pruckner N, Holthoff-Detto V. Antidepressant pharmacotherapy in old-age depression—a
review and clinical approach. Eur J Clin Pharmacol. 2017;73:661–7. https://doi.org/10.1007/
s00228-017-2219-1.
23. Bonin-Guillaume S, Benoit M. Depression. In: Roller-Wirnsberger R, Singler K, Polidori MC,
editors. Learning geriatric medecine. A study guide for medical students. Practical issues in
geriatrics. Cham: Springer Nature; 2018. p. 145–50.
24. Kivela SL, Viramo P, Pakhala K. Factors predicting the relapse of depression in old age. Int
J Geriatr Psychiatry. 2000;15:112–9. https://doi.org/10.1002/(sici)1099-1166(200002)15:2
<112::aid-gps83>3.0.co;2-b.
378 S. Bonin-Guillaume
25. Lu CY, Rougheaud E. New users of antidepressant medications: first episode duration and
predictors of discontinuation. Eur J Clin Pharmacol. 2012;68:65–71. https://doi.org/10.1007/
s00228-011-1087-3.
26. Braunstein D, Hardy A, Boucherie Q, Frauger E, Blin O, Gentile G, Micallef J. Antidepressant
adherence patterns in older patients: use of a clustering method on a prescription database.
Fundam Clin Pharmacol. 2017;31:226–36.
27. Bautista LE, Vera-Cala LM, Colombo C, Smith P. Symptoms of depression and anxiety and
adherence to antihypertensive medication. Am J Hypertens. 2012;25:505–11. https://doi.
org/10.1038//ajh.2011.256.
28. Breining A, Bonnet-Zamponi D, Zerah L, Micheneau C, Riolacci-Dhoyen N, Chan-Chee C,
et al. Exposure to psychotropics in the French older population living with dementia: a nation-
wide population-based study. Int J Geriatr Psychiatry. 2017;32:750–60.
29. Jacquin-Piques A, Sacco G, Tavassoli N, Rouaud O, Bejot Y, Giroud M, et al. Psychotropic
drug prescription in patients with dementia: nursing home residents versus patients living at
home. J Alzheimers Dis. 2016;49:671–80. https://doi.org/10.3233/JAD-150280.
30. Alexopoulos GS, Abrams RC, Young RC, Shamoian CA. Cornell scale for depression in
dementia. Biol Psychiatry. 1988;23:271–81.
31. Banerjee S, Hellier J, Dewey M, Romeo R, Ballard C, Baldwin R, et al. Sertraline or mirtazapine
for depression in dementia (HTA-SADD): a randomised, multicentre, double-blind, placebo-
controlled trial. Lancet. 2011;378:403–11. https://doi.org/10.1016/S0140-6736(11)60830-1.
32. Dudas R, Malouf R, McCleery J, Dening T. Antidepressants for treating depression in demen-
tia. Cochrane Database Syst Rev. 2018;(8):CD003944. https://doi.org/10.1002/14651858.
CD003944.pub2.
33. Lee HB, Lyketsos CG. Depression in Alzheimer disease: heterogeneity and related issues. Biol
Psychiatry. 2003;54:353–62. https://doi.org/10.1016/s0006-3223(03)00543-2.
34. Starkstein SE, Jorge R, Mizrahi R, Robinson RG. The construct of minor and major depres-
sion in Alzheimer’s disease. Am J Psychiatry. 2005;162:2086–93. https://doi.org/10.1176/
appi.ajp.162.11.2086.
35. Benoit M, Berrut G, Doussaint J, Bakchine S, Bonin-Guillaume S, Fremont P, et al. Apathy
and depression in mild Alzheimer’s disease: a cross-sectional study using diagnostic criteria. J
Alzheimers Dis. 2012;31:325–34.
36. Haute Autorité de Santé. Recommandation de bonnes pratiques. Maladie D’Alzheimer et
Maladies Apparentées : diagnostic et prise en charge de l’apathie. (Guidelines for Alzheimer’s
disease and related disorders: diagnosis and management of apathy) July 2014; 46p. www.has.
fr. Accessed 2 Nov 2021.
37. Sansone RA, Sansone LA. SSRI-induced indifference. Psychiatry. 2010;7:14–8.
38. Fotso Soh J, Klil-Drori S, Rej S. Using lithium in older age bipolar disorder: special consider-
ations. Drugs Aging. 2019;36:147–54. https://doi.org/10.1007/s40266-018-0628-1.
39. Gerhard T, Devanand DP, Huang C, Stephen CS, Olfson M. Lithium treatment and risk for
dementia in adults with bipolar disorder: population-based cohort study. Br J Psychiatry.
2015;207:46–51. https://doi.org/10.1192/bjp.bp.114.154047.
40. Wiles N, Thomas L, Abel A, Ridgway N, Turner N, Campbell J, et al. Cognitive behavioural
therapy as an adjunct to pharmacotherapy for primary care based patients with treatment resis-
tant depression: results of the CoBalT randomised controlled trial. Lancet. 2013;381:375–84.
https://doi.org/10.1016/S0140-6736(12)61552-9.
41. Kerner N, Prudic J. Current electroconvulsive therapy practice and research in the geriatric
population. Neuropsychiatry. 2014;4:33–54. https://doi.org/10.2217/npy.14.3.
42. Pinna M, Manchia M, Oppo R, Scano F, Pillai G, Loche AP, et al. Clinical and biological predic-
tors of response to electroconvulsive therapy (ECT): a review. Neurosci Lett. 2018;669:32–42.
43. Stek ML, van der Wurff FB, Uitdehaag BMJ, Beekman ATF, Hoogendijk WJG. ECT in the
treatment of depressed elderly: lessons from a terminated clinical trial. Int J Geriatr Psychiatry.
2007;22:1052–4. https://doi.org/10.1002/gps.1800.
44. Jalenques I, Legrand G, Vaille-Perret E, Tourtauchaux R, Galland F. Therapeutic efficacy and
safety of repetitive transcranial magnetic stimulation in depression of the elderly: a review.
Encéphale. 2010;36(Suppl 2):D105–18. https://doi.org/10.1016/j.encep.2009.10.007.
25 Optimizing Pharmacotherapy in Older Patients with Depression or Anxiety 379
26.1 Introduction
E. Kiesswetter (*)
Institute for Biomedicine of Aging, Friedrich-Alexander-Universität Erlangen Nürnberg,
Nürnberg, Germany
e-mail: [email protected]
C. C. Sieber
Institute for Biomedicine of Aging, Friedrich-Alexander-Universität Erlangen Nürnberg,
Nürnberg, Germany
Department of Medicine, Kantonsspital Winterthur, Winterthur, Switzerland
e-mail: [email protected]
Fig. 26.1 The DoMAP schematic model illustrating the various factors contributing to malnutri-
tion in older people. Level 1 in dark green represents the core etiological mechanisms that lead to
malnutrition. Level 2 in light green consists of the factors that lead directly to one or more of the
core etiological mechanisms that cause malnutrition. Level 3 in yellow lists the various common
conditions that indirectly contribute to malnutrition [18]
Even though there is broad consensus by experts on the multiple causes of mal-
nutrition in older adults, evidence for many of these determinants is low or conflict-
ing [19, 20]. This is mainly due to a different operationalization of determinants and
malnutrition among studies, inappropriate confounder control, and lack of longitu-
dinal or randomized trials.
The European Society for Clinical Nutrition and Metabolism (ESPEN) recom-
mends the Mini Nutritional Assessment (MNA) as screening tool for malnutrition
in older people [22] as the MNA was specifically developed for older people and
can be used in the outpatient and institutional settings [23]. Its short form has six
items comprising the core aspects of reduced food intake, weight loss, acute
stress, and BMI. In addition, there are questions on mobility limitations and neu-
ropsychological problems—two important risk factors for malnutrition in older
people. A further feature is the option to exchange the BMI item with calf circum-
ference if BMI assessment is not possible, e.g., due to immobility. According to a
sum score ranging from 0 to 14 points, patients are categorized as well-nourished
(12–14 points), at risk of malnutrition (8–11 points), or malnourished (<7
points) [24].
A screening alternative in the hospital setting is the Nutritional Risk Screening
tool (NRS) [25], specifically developed for the acute care setting and which can
be used also in younger patients. The NRS is designed to identify patients who
might benefit from nutrition interventions. It is composed of a prescreening with
four questions on the core aspects low BMI, recent weight loss, reduced dietary
intake, and severe disease. In case of a positive prescreening, the above core ele-
ments are quantified during the main screening. The age of patients (≥70 years) is
considered as a further risk factor. Three or more points on the NRS indicate
nutritional risk.
There are several other screening tools available that are less often used in clinical
practice. In a systematic review on the validity of malnutrition screening instruments
in older people, 34 different tools that have been used in older people were identified,
of which 23 were specifically developed for this age group [26]. The usage of a spe-
cific tool often depends on the health policies and guidelines of the respective
countries.
26 Nutritional Deficiency and Malnutrition 385
Following a positive screening result, the nutritional status of an older person needs
to be further assessed to form the basis for targeted interventions aiming to improve
nutritional status, the overall clinical course of the presenting illness, and the quality
of life [21].
Both, in research and clinical practice, different criteria and cutoff values are used
for the diagnosis of malnutrition. To align the diagnosis of malnutrition, a coalition
of several international medical nutrition societies mostly from Europe, the USA,
and Asia—the so-called Global Leadership Initiative on Malnutrition (GLIM)—
compiled consensus-based criteria for malnutrition [27]. The concept proposes that
after a positive screening, the phenotypic criteria “unintentional weight loss,” “low
BMI,” and “low muscle mass” as well as the etiologic criteria “reduced food intake
or assimilation” and “inflammation” should be assessed (Table 26.1). If at least one
phenotypic and one etiological criterion apply, malnutrition can be assumed. The
severity of malnutrition can be further evaluated based on the expression of the
phenotypic criteria [27].
In clinical trials, muscle mass or lean body mass and fat-free mass as its proxies
are usually assessed by bioelectrical impedance analysis (BIA) or dual-energy
Table 26.1 Phenotypic and etiological criteria to diagnose malnutrition (based on Global
Leadership Initiative [27])
Phenotypic criteria
Weight loss >5% within past 6 months
or
>10% beyond 6 months
Low body mass index <20 kg/m2, if <70 years
or
<22 kg/m2, if ≥70 years
Reduced muscle mass Depends on the measuring technique,
e.g., BIA: FFMI <17 (m), <15 (f) kg/m2,
BIA, DXA: ASMI <7 (m), <6 (f) kg/m2
Etiologic criteria
Reduced food intake or ≤50% of energy needs >1 week
assimilation or
any reduction for >2 weeks
or
any chronic gastrointestinal condition adversely affecting food
assimilation or absorption, e.g., short bowel syndrome or chronic
diarrhea
Inflammation Acute disease/injury, e.g., infections, burns, trauma
Chronic diseases, e.g., cancer, chronic obstructive pulmonary disease
BIA Bio-electrical impedance analysis, DXA Dual-energy X-ray absorptiometry, FFMI Fat free
mass index, ASMI Appendicular skeletal muscle mass index, m male, f female
386 E. Kiesswetter and C. C. Sieber
X-ray absorptiometry (DXA). As these methods are often not available in routine
clinical practice or are too expensive, calf circumference is often used as an indica-
tor of muscle mass (cutoff <31 cm for both sexes).
By quantifying the nutritional deficits, the necessity for and the amount of nutri-
tional interventions can be determined. The fundamental metrics for the determina-
tion are the individual’s energy and protein intake and the corresponding
requirements.
As in routine clinical practice, measurement of energy requirements is usually
not possible, formulae to estimate resting energy expenditure, e.g., by Mueller [28]
or Harris-and Benedict [29], are used. For this purpose, details of age, sex, and body
weight are needed. In combination with the physical activity level (PAL), total
energy expenditure can be calculated. A PAL between 1.2 and 1.3 represents a low
energy expenditure in frail, immobile, or bedridden older people. For older people,
a guiding value for energy intake of 30 kcal/kg body weight (BW)/day is proposed,
which needs to be adjusted according to nutritional status, physical activity level,
disease status, and tolerance [21].
Protein requirements of older people are currently the subject of the much scientific
debate, and reference values differ by country. While the European Food Safety
Authority recommends an intake of 0.8 g/kg BW/day for young and older adults [30],
expert groups suggest a higher optimal protein intake in older people to prevent decline
in muscle mass (1–1.2 g/kg BW/day) [31]. Higher amounts might be needed to com-
pensate for increased metabolic demands due to inflammation, sepsis, or wounds.
Intake can be assessed by dietary records of at least 3 days, where all consumed
foods and their quantities are described in detail. An approximate measure of the
amount eaten is plate diagrams where the intake is estimated by quarters of the
offered amount [nothing, ¼, ½, ¾, all].
Based on the nutritional assessment, realistic and individualized goals for nutri-
tional therapy need to be established, considering potential risks, contraindications,
benefits as well as resources, needs, and preferences of the older patient. Nutritional
interventions generally incorporate the following options:
26.6 Conclusions
Nutritional deficiency and malnutrition are common in older people in all care set-
tings and are commonly multifactorial. Early detection by a routine screening, a
thorough comprehensive nutritional assessment, and an individualized and multi-
disciplinary therapy regimen can improve nutritional status, related clinical out-
comes, and overall prognosis in older people with malnutrition.
388 E. Kiesswetter and C. C. Sieber
References
1. Allison SP. Malnutrition, disease, and outcome. Nutrition. 2000;16(7–8):590–3.
2. Sharkey JR, Branch LG, Zohoori N, Giuliani C, Busby-Whitehead J, Haines PS. Inadequate
nutrient intakes among homebound elderly and their correlation with individual characteristics
and health-related factors. Am J Clin Nutr. 2002;76(6):1435–45.
3. Rosenberger C, Rechsteiner M, Dietsche R, Breidert M. Energy and protein intake in 330
geriatric orthopaedic patients: are the current nutrition guidelines applicable? Clin Nutr
ESPEN. 2019;29:86–91.
4. de Groot CP, van Staveren WA. Nutritional concerns, health and survival in old age.
Biogerontology. 2010;11(5):597–602.
5. Hengeveld LM, Boer JMA, Gaudreau P, Heymans MW, Jagger C, Mendonça N, et al.
Prevalence of protein intake below recommended in community-dwelling older adults: a
meta-analysis across cohorts from the PROMISS consortium. J Cachexia Sarcopenia Muscle.
2020;11(5):1212–22.
6. Kehoe L, Walton J, Flynn A. Nutritional challenges for older adults in Europe: current status
and future directions. Proc Nutr Soc. 2019;78(2):221–33.
7. ter Borg S, Verlaan S, Hemsworth J, Mijnarends DM, Schols JM, Luiking YC, et al.
Micronutrient intakes and potential inadequacies of community-dwelling older adults: a sys-
tematic review. Br J Nutr. 2015;113(8):1195–206.
8. Tieland M, Borgonjen-Van den Berg KJ, Van Loon LJ, de Groot LC. Dietary protein intake in
Dutch elderly people: a focus on protein sources. Nutrients. 2015;7(12):9697–706.
9. Cereda E, Pedrolli C, Klersy C, Bonardi C, Quarleri L, Cappello S, et al. Nutritional status in
older persons according to healthcare setting: a systematic review and meta-analysis of preva-
lence data using MNA(R). Clin Nutr. 2016;35(6):1282–90.
10. Marshall S, Bauer J, Isenring E. The consequences of malnutrition following discharge from
rehabilitation to the community: a systematic review of current evidence in older adults. J Hum
Nutr Diet. 2014;27(2):133–41.
11. Correia MI, Waitzberg DL. The impact of malnutrition on morbidity, mortality, length of hospi-
tal stay and costs evaluated through a multivariate model analysis. Clin Nutr. 2003;22(3):235–9.
12. Norman K, Pichard C, Lochs H, Pirlich M. Prognostic impact of disease-related malnutrition.
Clin Nutr. 2008;27(1):5–15.
13. Lorenzo-López L, Maseda A, de Labra C, Regueiro-Folgueira L, Rodríguez-Villamil JL,
Millán-Calenti JC. Nutritional determinants of frailty in older adults: a systematic review.
BMC Geriatr. 2017;17(1):108.
14. Ligthart-Melis GC, Luiking YC, Kakourou A, Cederholm T, Maier AB, de van der Schueren
MAE. Frailty, sarcopenia, and malnutrition frequently (co-)occur in hospitalized older adults:
a systematic review and meta-analysis. J Am Med Dir Assoc. 2020;21(9):1216–28.
15. Hai S, Gao Q, Gwee X, Chua DQL, Yap KB, Ng TP. Malnutrition risk, physical function
decline and disability in middle-aged and older adults followed up in the Singapore longitudi-
nal ageing study. Clin Interv Aging. 2021;16:1527–39.
16. Buurman BM, Hoogerduijn JG, de Haan RJ, Abu-Hanna A, Lagaay AM, Verhaar HJ, et al.
Geriatric conditions in acutely hospitalized older patients: prevalence and one-year survival
and functional decline. PLoS One. 2011;6(11):e26951.
17. Rasheed S, Woods RT. Malnutrition and quality of life in older people: a systematic review and
meta-analysis. Ageing Res Rev. 2013;12(2):561–6.
18. Volkert D, Kiesswetter E, Cederholm T, Donini LM, Eglseer D, Norman K, et al. Development
of a model on determinants of malnutrition in aged persons: a MaNuEL project. Gerontol
Geriatr Med. 2019;5:2333721419858438.
19. van der Pols-Vijlbrief R, Wijnhoven HA, Schaap LA, Terwee CB, Visser M. Determinants
of protein-energy malnutrition in community-dwelling older adults: a systematic review of
observational studies. Ageing Res Rev. 2014;18:112–31.
26 Nutritional Deficiency and Malnutrition 389
20. O'Keeffe M, Kelly M, O'Herlihy E, O'Toole PW, Kearney PM, Timmons S, et al. Potentially
modifiable determinants of malnutrition in older adults: a systematic review. Clin Nutr.
2019;38(6):2477–98.
21. Volkert D, Beck AM, Cederholm T, Cruz-Jentoft A, Goisser S, Hooper L, et al. ESPEN guide-
line on clinical nutrition and hydration in geriatrics. Clin Nutr. 2019;38(1):10–47.
22. Kondrup J, Allison SP, Elia M, Vellas B, Plauth M. ESPEN guidelines for nutrition screening
2002. Clin Nutr. 2003;22(4):415–21.
23. Guigoz Y, Vellas B, Garry PJ. Mini Nutritional Assessment: A practical assessment tool for
grading nutritional state of elderly patients. Facts Res Gerontol. 1994;(Supplement 2):15–59.
24. Kaiser MJ, Bauer JM, Ramsch C, Uter W, Guigoz Y, Cederholm T, et al. Validation of the mini
nutritional assessment short-form (MNA-SF): a practical tool for identification of nutritional
status. J Nutr Health Aging. 2009;13(9):782–8.
25. Kondrup J, Rasmussen HH, Hamberg O, Stanga Z. Nutritional risk screening (NRS 2002): a
new method based on an analysis of controlled clinical trials. Clin Nutr. 2003;22(3):321–36.
26. Power L, Mullally D, Gibney ER, Clarke M, Visser M, Volkert D, et al. A review of the validity
of malnutrition screening tools used in older adults in community and healthcare settings—a
MaNuEL study. Clin Nutr ESPEN. 2018;24:1–13.
27. Cederholm T, Jensen GL, Correia M, Gonzalez MC, Fukushima R, Higashiguchi T, et al.
GLIM criteria for the diagnosis of malnutrition—a consensus report from the global clinical
nutrition community. Clin Nutr. 2019;38:1–9.
28. Müller MJ, Bosy-Westphal A, Klaus S, Kreymann G, Lührmann PM, Neuhäuser-Berthold M,
et al. World Health Organization equations have shortcomings for predicting resting energy
expenditure in persons from a modern, affluent population: generation of a new reference stan-
dard from a retrospective analysis of a German database of resting energy expenditure. Am J
Clin Nutr. 2004;80(5):1379–90.
29. Harris JA, Benedict FG. A biometric study of human basal metabolism. Proc Natl Acad Sci U
S A. 1918;4(12):370–3.
30. European Food Safety Authority EPoDP, Nutrition and Allergies (NDA). Scientific opinion on
dietary reference values for protein. EFSA J. 2012;10:1–66.
31. Bauer J, Biolo G, Cederholm T, Cesari M, Cruz-Jentoft AJ, Morley JE, et al. Evidence-based
recommendations for optimal dietary protein intake in older people: a position paper from the
PROT-AGE Study Group. J Am Med Dir Assoc. 2013;14(8):542–59.
Pharmacotherapy of Insomnia in Older
Adults 27
Mirko Petrovic
Separately from age-related changes, sleep problems affect 10–15% of the general
population and increase with older age as well [1, 2]. Sleep disturbances include
insomnia, as the most common sleep disorder and therefore the most researched
one, dyssomnia (restless legs, periodic limb movement, sleep apnoea, narcolepsy)
and parasomnia (REM sleep behaviour disorder, night terrors, sleepwalking).
Insomnia is often identified as the subjective complaint of problems with initiating
or maintaining sleep, or non-restorative sleep, causing significant daytime symp-
toms such as concentration problems and mood disturbances. The overall preva-
lence of insomnia symptoms ranges from 30% to 48% among older adults [3–6],
while the prevalence of insomnia disorder ranges from 12% to 20% [7]. Insomnia is
usually categorised by the predominant symptom of either difficulty in sleep onset
or sleep maintenance. Sleep maintenance symptoms are most prevalent among
adults complaining of insomnia (50–70%), followed by difficulty in initiating sleep
(35–60%) and non-restorative sleep (20–25%). An incidence rate for insomnia
symptoms of 5% per year, with a yearly incidence of 8% at 1-year follow-up, has
been observed among older adults [8, 9].
27.2 Treatment
M. Petrovic (*)
Section of Geriatrics, Department of Internal Medicine and Paediatrics, Ghent University,
Ghent, Belgium
e-mail: [email protected]
interventions for insomnia in adults of all ages. For the purpose of this chapter,
however, only the pharmacological options have been discussed here in detail.
BZDs and Z-drugs are the most commonly used symptomatic treatment for insom-
nia. The term benzodiazepine has been related to the portion of the structure com-
posed of a benzene ring fused to a seven-membered diazepine ring. Since almost all
BZDs contain a 5-aryl substituent and a 1,4 diazepine ring, the term actually denotes
the 5-aryl-1,4-benzodiazepines. Although variations in the structure of the ring sys-
tems yield different chemical compounds, pharmacological activity remains similar
[29, 30]. Three non-benzodiazepine receptor agonists are now available: zaleplon (a
pyrazolopyrimidine), zolpidem (an imidazopyridine) and zopiclone (a cyclopyrro-
lone). Although termed non-BZD, Z-drugs interact with BZD receptors [14–16].
Almost all the effects of the BZDs result from actions on the central nervous
system (CNS).
Molecular targets for BZD actions in the CNS are inhibitory neurotransmitter
receptors directly activated by the amino acid gamma-aminobutyric acid (GABA)
[14, 15].
Two central BZD receptor subtypes (BZ1, and BZ2,) and one peripheral BZD
receptor have been categorised. BZ1, (also called omega 1) receptors are located in
the cerebellum. Hypnosedative and anxiolytic actions are mediated mainly through
the BZ1 receptor subtype. BZ2, (omega 2) receptors, on the other hand, are located
predominantly in the spinal cord and striatum. These receptors may be involved in
mediating the muscle relaxant actions of BZDs [17]. Most BZDs interact non-
selectively with both receptor subtypes, which result in a variety of inhibitory cen-
tral nervous system effects [18]. The peripheral BZD receptor is located in the
kidney. Its role in anxiolytic and hypnosedative actions remains unclear [17].
There are obvious differences in potency between different BZDs. The equiva-
lent dose may differ as much as 20-fold [19, 20]. We should keep this in mind when
substituting one BZD by another. These differences in potency relate to differences
in affinity for various receptor subtypes.
27 Pharmacotherapy of Insomnia in Older Adults 393
Almost all of the BZDs are completely absorbed; some of them reach the sys-
temic circulation only in the form of active metabolites. As a result, three categories
BZDs are identified based on the elimination half-lives (t1/2): short-acting agents (t1/2
2–5 h), intermediate acting agents (t1/2 6–24 h) and long-acting agents (t1/2 more than
24 h) [15].
The BZDs and their metabolites have a high affinity for binding to plasma pro-
teins. The degree of binding correlates with lipid solubility and varies from 70 to 99
%. Most BZDs have a large volume of distribution due to their high lipid solubility.
The concentration in cerebrospinal fluid is practically the same as the concentration
of free drug in plasma [21].
BZDs are metabolised in the liver primarily by oxidation, nitroreduction and
glucuronidation [15, 22]. Most of them can be classified as low clearance drugs.
Alterations in BZD pharmacokinetics have been observed in older adults. A pro-
longation of the half-life of the oxidised drugs has been reported in this age group,
but the clearance or half-life of the glucuronised drugs is less affected. Consequently,
the BZDs metabolised by oxidation (e.g. flurazepam and diazepam) should not be
prescribed to older adults, since higher plasma concentrations for a given drug dos-
age and consequently enhanced clinical effects are expected [21, 23].
Other significant and often seen age-related changes that may alter BZD metabo-
lism include decreased liver blood flow, plasma albumin and lean body mass.
Reduced hepatic blood flow can change the plasma concentration-time profile and
increase peak concentrations. Decrease in plasma albumin levels may affect protein-
binding capacity as well. Distribution volume may increase as a consequence of
decreased lean body mass and increased proportion of fat. This results in an
increased elimination half-time, prolonged effects after administration and accumu-
lation of active metabolites [24, 25].
Changes in pharmacodynamics may also occur, as a result of altered affinity of
the receptors, which implies a greater effect for a given plasma concentration. Older
adults are more vulnerable to the effects of BZDs on cognitive function, especially
at higher dosages. Different researchers report that older adults require both a lower
dose and a lower plasma concentration to cause a constant level of sedation [26, 27].
Memory loss, altered balance and high risk of falls and fractures have been reported
after short-term administration of BZDs [28, 29].
All BZDs are characterised by, in somewhat altering degrees, five major effects:
hypnosedative, anxiolytic, anticonvulsant, muscular relaxant and amnesic. In the
short term, BZDs may be used safely in certain clinical conditions. With long-term
use, tolerance, dependence and withdrawal effects may become major drawbacks
[30, 31].
As to hypnotic effects, BZDs accelerate sleep onset, decrease nocturnal arousals
and increase total sleep time [23]. However, they change the normal sleep pattern:
light sleep is prolonged, while the duration of restorative slow wave sleep and rapid
394 M. Petrovic
eye movement sleep is reduced. The onset of the first rapid eye movement sleep
episode may be delayed [20]. Although some long-term users of BZDs are self-
assured that sleep quality and duration are bettered as a result of the treatment, the
findings of polysomnographic studies illustrate that the sleep pattern remains clearly
different from normal sleep [32]. The aberrant sleep profile possibly results from
unselective depression of both arousal and sleep mechanisms in the brainstem
[33, 34].
Anxiolytic effects are induced by doses that cause minimal sedation, although the
hypnotic, muscular relaxant and amnesic actions may all provide relief of associ-
ated tension and insomnia [35]. The effect on anxiety is presumably related to sup-
pressive activity in limbic and other brain areas involved in the development of
anxiety. The principal clinical characteristic of BZDs prescribed for anxiety is the
rapid onset of action, usually noticeable after a single dose. BZDs provide only
symptomatic treatment for anxiety. However, they might be indicated in the initial
management of distressing anxiety while awaiting enduring clinical effects from
more specific non-pharmacological treatments [17].
As to anticonvulsant effects, BZDs are effective in the treatment of status epilep-
ticus and convulsions due to drug poisoning and, however, can only be used in
emergency situations and are not indicated for the long-term treatment of epilepsy
because of the development of tolerance in the majority of patients [36, 37].
Muscular relaxant effects of BZDs can sometimes be used in case of different
motor disorders (i.e. dystonias and involuntary movements, myoclonus, restless
limbs syndrome and muscle spasm associated with pain) [38].
As to amnesic effects, BZDs also cause dose-related anterograde amnesia. These
amnesic effects may be clinically significant, particularly in older adults and in
those with coexisting medical problems [23, 39].
Older adults in particular are vulnerable to adverse effects of BZDs. These patients
are more sensitive to CNS depression, states of confusion and ataxia that can result
in falls and fractures [29, 40]. They are also prone to respiratory depression,
decreased ventilatory response, hypercapnia and increased hypopnoeic episodes
during sleep [40].
In addition to tolerance to the hypnotic and anxiolytic effects, ageing increases the
susceptibility to the side effects of BZDs. The atrophy of neurons and the increased
sensitivity of the CNS, together with pharmacokinetic changes, which lead to BZD/
Z-drug accumulation, increase the possibility of confusion and CNS side effects,
hangover effects and subsequent risk of falling [41–43]. Often falls have no major
consequences, but in frail older adults, they can cause injuries and fractures, leading
27 Pharmacotherapy of Insomnia in Older Adults 395
to hospitalisation and serious disability [44]. It has been questioned whether there is
an independent association between BZD/Z-drug use and increased fall risk. Not
only have falls a multifactorial aetiology but other psychotropic drugs are also asso-
ciated with a 30–70% increased risk of falls [45–47].
Even though BZDs at first induce and prolong sleep, tolerance develops shortly.
Sleep latency and duration regress to pre-treatment levels after a few weeks of con-
tinued treatment. Sleep quality, on the other hand, does not improve, since deep
NREM sleep and REM sleep stages are partially replaced by stage 2 light NREM
sleep [23, 48]. Tolerance to the anxiolytic properties of BZDs develops more gradu-
ally than to the hypnotic effects.
Rebound insomnia refers to an increase in the initial symptom beyond the base
level after withdrawal from BZDs. Population surveys and results from large treat-
ment effectiveness studies show rebound insomnia in 14–20% of patients treated
with BZDs, a rate indistinguishable from that seen with over-the-counter drugs or
placebo [49].
questionnaires) was more optimistic [55]. This indicates that there may be an over-
estimation of the BZD’s effects on sleep outcome parameters. Moreover, the selec-
tive reporting and publishing of positive studies and results also indicate that there
is an overestimation favouring the BZD/Z-drugs’ effects [56, 59]. In addition to the
possible overestimation of the BZD/Z-drug’s effects, several meta-analyses showed
that placebo treatment also improves the sleep significantly [60, 61]. Because
BZDs/Z-drugs are not free of side effects, the overall risk/benefit balance must be
taken into account. Moreover, all these studies investigated the effect of a short-term
treatment. The maximum study duration for BZDs is 8 weeks [62], and for Z-drugs,
it is 1 year [63, 64]. Although the majority of BZD/Z-drug users are chronic users,
research on long-term BZD or Z-drug use is lacking.
So far, there is a lack of a sound evidence base for the long-term efficacy of
BZD/Z-drug use. Because of practical barriers but also because the fact that long-
term BZD/Z therapy is not concordant with the guidelines, it is difficult to initiate
randomised controlled trials with incident users and follow the sleep longitudinal.
The studies that tried to evaluate long-term effectiveness found a worse sleep qual-
ity in long-term users [65–67]. After taking into consideration both the short-term
and long-term risks of BZD/Z-drug use, and in the light of questionable effect of
these drugs on insomnia in the long term, guidelines discourage chronic BZD/Z--
drug use.
This age group, with a higher risk of abovementioned side effects, needs tailored
prescribing which also includes deprescribing [68]. Discontinuation of BZDs/Z-
drugs is widely advised but has not reached global acceptance among prescribers,
caregivers and patients.
There is a general agreement on the notion that most patients who take BZDs
regularly should try and discontinue treatment [69, 70]. Many patients who have
taken BZDs for years can have these drugs withdrawn successfully. Men under the
age of 50 without personality disturbance show particularly good response after
withdrawal from BZDs. On the contrary, older women with anxious symptoms and
personality disturbance respond less successfully to withdrawing [71].
The factors presumed to affect withdrawal are personality profile, dose and half-
life of BZDs, duration of treatment and mode of withdrawal [71, 72]. On the con-
trary, there are reports suggesting that with the exception of age, withdrawal
outcome is not related to any particular variable [72].
Withdrawal symptoms, if any, occur most often after 3 months of habitual use.
They may include anxiety, restlessness, sleep disturbance, headache, muscle
cramps, nausea, delirium or convulsions.
The treatment of BZD withdrawal symptoms includes appropriate psychological
support together with a gradual dosage tapering. The level of psychological support
may range from simple encouragement to anxiety management or behavioural ther-
apy. Chronic insomnia can be effectively treated in older subjects with structured
27 Pharmacotherapy of Insomnia in Older Adults 397
and sleep-focused interventions designed to change poor sleep habits [73]. In most
of the cases, sleep symptoms progressively improve after withdrawal. It has been
demonstrated that older adults tolerate withdrawal as well as if not better than young
individuals [74].
There are no unanimous recommendations in the literature with regard to the
optimal duration of the withdrawal process. Therefore, a variable withdrawal period
ranging from 2 to 12 weeks may be allowed. The size of the stepwise lowering in
dosage that should be applied is arguable as well. Some researchers propose a fixed
tapering schedule, while others claim that the reduction rate should be titrated
against the patient’s withdrawal symptoms [75].
In a systematic review of interventions to deprescribe BZDs among older adults,
seven studies of benzodiazepines and Z-drug withdrawal were identified.
Benzodiazepine discontinuation rates were 64.3% in one study that applied pharma-
cological substitution with melatonin and 65.0% in a study that applied general
practitioner-targeted intervention. Mixed interventions including patient education
and tapering (n = 2), pharmacological substitution with psychological support
(n = 1) and tapering with psychological support (n = 1) yielded discontinuation rates
between 27.0 and 80.0%. Five studies measured clinical outcomes following benzo-
diazepine discontinuation. Most of the studies (n = 4) observed no difference in
prevalence of withdrawal symptoms or sleep quality, while one study reported
decline in quality of life in those who continued taking benzodiazepine vs. those
who discontinued over 8 months [76].
Most older BZD/Z users for the indication insomnia are low-dose users. Although
there is not much research done on which BZD/Z-drug is more resistant to with-
drawal, generally, short-acting BZD/Z-drugs are more likely to cause withdrawal
symptoms upon discontinuation than long-acting BZDs. Therefore, substitution
with a long-acting BZD (diazepam) can lessen (and delay) withdrawal symptoms
[77]. However, it is not appropriate to use long-acting BZDs/Z-drugs in older adults
(in concordance with all explicit criteria). The long half-life can result in accumula-
tion. Therefore, substitution with an intermediate working BZD/Z-drug (lorazepam/
lormetazepam) or simply stay with the same product when discontinuing is advised.
Psychological interventions which support withdrawal, such as cognitive behav-
ioural therapy (CBT), have shown to be effective, also in older adults [78, 79].
These psychological techniques are intended to facilitate withdrawal and motivate
patients to maintain abstinent over time (relapse prevention).
A meta-analysis focusing on older adults showed that the best strategy for dis-
continuing BZDs/Z-drugs in older adults residing in the nursing home setting was
the combination of supervised withdrawal with psychotherapy [80]. Supervising the
withdrawal in a setting with supporting care personnel is possible, but delivering
psychotherapy seems more difficult to implement. A promising strategy for reduc-
ing BZDs/Z-drugs in the nursing home setting was the implementation of a pharma-
cist medication review [81, 82]. Often, the pharmacists gave recommendations or
feedback to the nursing staff and delivered “an educational message”. Also, a pro-
gramme led by a psychologist which educated prescribers and caregivers, offered
health education and relaxation training to the residents, reduced the overall
398 M. Petrovic
prevalence of BZD/Z-drug use (from 70% to 35%) in this setting [83]. The educa-
tional tools used in long-term care facilities varied from prescribing algorithms and
bulletins to staff meetings and lectures [84]. However, educational interventions
alone cannot be expected to change behaviour when healthcare providers do not
perceive it to be important, or when the change is complex and dependent on the
interaction of too many stakeholders.
Several studies have investigated BZD/Z-drug withdrawal in the nursing home
setting and focused on the success rate (focus on residents already willing to discon-
tinue) [83, 85, 86]. However, health policy-makers should have an idea of the extent
of the target population and the overall feasibility, including the willingness towards
discontinuation in this setting. Therefore, general practitioner, nurse and, more
importantly, the patient have to be susceptible towards change. Moreover, a lack of
knowledge on non-pharmacological treatments and a lack of their availability and/
or time to administer make it difficult for health providers to offer alternatives when
withdrawal or rebound symptoms emerge. Some studies show that patients are sus-
ceptible towards change. Therefore, it is advised to routinely raise the issue of dis-
continuation and negotiate dose reduction [87, 88]. Current evidence shows that
benzodiazepine withdrawal is feasible in older population, but withdrawal rates
vary according to the type of intervention. As the benefits and sustainability of these
interventions are unclear, further studies should be conducted to assess this.
There are several pharmacotherapeutic alternatives that are used as alternative for
BZDs including antidepressants, melatonin receptor agonists and orexin receptor
antagonists. Different antidepressants, including trazodone, tricyclic antidepres-
sants (doxepin) and serotonergic antidepressant (mirtazapine), have sedating prop-
erties and are often used for the treatment of insomnia [3].
However, despite the fact that these alternatives have been shown to be more
effective than placebo at bettering short-term sleep outcomes, the size of effect is
variable; most trials have been industry sponsored, posing questions about publica-
tion bias, and the overall estimation of risk-to-benefit ratio is low [13]. The potential
benefits of pharmacotherapy on sleep quality and daytime function should always be
balanced against the risk of side effects and physical and psychological dependence
with long-term use.Trazodone is often prescribed for insomnia in doses of 25–100 mg.
A study on trazodone comparing its effect with zolpidem in 21- to 65-year-olds
showed a similar efficacy for sleep latency and sleep efficiency; however, both effects
decrease after the first week [89]. Adverse effects such as cardiac arrhythmias, ortho-
static hypotension, dizziness and potential priapism are well documented and can be
important in older population [90]. There is lack of evidence about pharmacotherapy
of sleep disorders in older adults with dementia. Several small trials showed certain
beneficial effects on sleep outcomes from trazodone, although larger trials are needed
to allow more sound conclusions. Systematic assessment of adverse effects in future
studies is essential [91]. However, clinical practice guidelines from the American
27 Pharmacotherapy of Insomnia in Older Adults 399
Academy of Sleep Medicine suggest that trazodone should not be used for treatment
of insomnia because its harms prevail over its benefits [13].
Doxepin is the only antidepressant approved by the Food and Drug Administration
(FDA) for insomnia at doses of 3–6 mg. Studies of adults aged 65 years and older
have shown, based on patient reports, that doxepin in doses of 1 mg and 3 mg,
respectively, improved measures of sleep onset, sleep duration, sleep quality and
global treatment outcomes over the 12-week study period [92].
Mirtazapine, a serotonergic antidepressant, might also improve insomnia. In a
study of adults with mean age of 40.9 years, the mirtazapine group showed improve-
ment in sleep latency, sleep efficiency and awakenings after sleep onset after
2 weeks of treatment. However, because of conflicting evidence and tolerance to its
sedative effects, it should not be used to treat insomnia in the absence of depres-
sion [93].
Ramelteon is a melatonin receptor agonist. It is approved by the FDA for treat-
ment of insomnia. In a study of older adults, ramelteon reduced patient reports of
sleep latency over 5 weeks of treatment with no significant rebound insomnia or
withdrawal effects [94, 95].
Melatonin at a dose of 2 mg has been approved by the European Medicines
Agency (EMA) for the short-term treatment of primary insomnia in patients aged
55 years and older. However, formal recommendations for the use of melatonin in
the treatment of insomnia still require further research [13, 96, 97].
Suvorexant is an orexin receptor antagonist. It is the first FDA-approved dual
orexin receptor antagonist and may be prescribed up to a 20-mg dose. It targets
wakefulness-promoting neuropeptides that regulate the sleep-wake cycle. It has
shown to be effective in decreasing sleep latency and in increasing total sleep time.
Suvorexant has been studied in both older (age 65 years or older) and younger (age
18–64 years) patients, showing no significant differences efficacy or safety between
these two groups. Long-term data are still lacking [98–100].
27.11 Conclusions
References
1. Conn D, Madan R. Use of sleep-promoting medications in nursing home residents: risks
versus benefits. Drugs Aging. 2006;23:271–87.
2. Wolkove N, Elkholy O, Baltzan M, Palayew M. Sleep and aging: 1. Sleep disorders com-
monly found in older people. Can Med Assoc J. 2007;176:1299–304.
3. Patel D, Steinberg J, Patel P. Insomnia in the elderly: a review. J Clin Sleep Med.
2018;14:1017–24.
4. Crowley K. Sleep and sleep disorders in older adults. Neuropsychol Rev. 2011;21:41–53.
5. Ohayon M. Epidemiology of insomnia: what we know and what we still need to learn. Sleep
Med Rev. 2002;6(2):97–111.
6. Hillman D, Murphy A, Pezzullo L. The economic cost of sleep disorders. Sleep.
2006;29:299–305.
7. Roth T, Coulouvrat C, Hajak G, Lakoma M, Sampson N, Shahly V, Shillington A, Stephenson
J, Walsh J, Kessler R. Prevalence and perceived health associated with insomnia based
on DSM-IV-TR; International Statistical Classification of Diseases and Related Health
Problems, Tenth Revision; and Research Diagnostic Criteria/International Classification of
Sleep Disorders, Second Edition criteria: results from the America Insomnia Survey. Biol
Psychiatry. 2011;69:592–600.
8. Foley D, Monjan A, Simonsick E, Wallace R, Blazer D. Incidence and remission of insom-
nia among elderly adults: an epidemiologic study of 6,800 persons over three years. Sleep.
1999;22(Suppl 2):S366–72.
9. Gureje O, Oladeji B, Abiona T, Lebowitz M. The natural history of insomnia in the Ibadan
study of ageing. Sleep. 2011;34:965–73.
10. Naylor E, Penev P, Orbeta L, Janssen I, Ortiz R, Colecchia E, Keng M, Finkel S, Zee P. Daily
social and physical activity increases slow wave sleep and daytime neuropsychological per-
formance in the elderly. Sleep. 2000;23:87–95.
11. McCurry S, Logsdon R, Teri L, Vitiello M. Evidence-based psychological treatments for
insomnia in older adults. Psychol Aging. 2007;22:18–27.
12. Smith M, Perlis M, Park A, Smith M, Pennington J, Giles D, Buysse D. Comparative meta-
analysis of pharmacotherapy and behavior therapy for persistent insomnia. Am J Psychiatry.
2002;159:5–11.
13. Sateia M, Buysse D, Krystal A, Neubauer D, Heald J. Clinical practice guideline for the phar-
macologic treatment of chronic insomnia in adults: an American Academy of Sleep Medicine
clinical practice guideline. J Clin Sleep Med. 2017;13:307–49.
14. Petrovic M, Mariman A, Warie H, Afschrift M, Pevernagie D. Is there a rationale for pre-
scription of benzodiazepines in the elderly? Review of the literature. Acta Clin Belg.
2003;58:27–36.
15. Hobbs W, Rail T, Verdoom T. Hypnotics and sedatives; ethanol. In: Hardman JG, Gilman
AG, Limbird LE, editors. Goodman & Gilman’s the pharmacological basis of therapeutics.
New York: Me Graw-Hill Health Profession Division; 1996. p. 361–97.
16. Heydom W. Zaleplon—a review of a novel sedative hypnotic used in the treatment of insom-
nia. Exp Opin Invest Drugs. 2000;9:841–58.
17. Stahl S. Anxiolytic and sedative-hypnotics. In: Stahl SM, editor. Essential psychopharmacol-
ogy. Cambridge: Cambridge University Press; 2000. p. 297–333.
18. Kirkwood CK. Management of insomnia. J Am Pharm Assoc. 1999;39:688–96.
19. Roth T, Roehrs TA. Issues in the use of benzodiazepine therapy. J Clin Psychiatry 1992; 53
(Suppl 6): 14–18.
20. Ashton H. Guidelines for rational use of benzodiazepines -when and what to use. Drugs.
1994;48:25–40.
21. Hammerlein A, Derendorf H, Lowenthal D. Pharmacokinetic and pharmacodynamic changes
in the elderly. Clin Pharmacokinet. 1998;35:49–64.
22. Breiner D. Clinical pharmacokinetics of hypnotics. Clin Pharmacokinet. 1997;2:93–109.
27 Pharmacotherapy of Insomnia in Older Adults 401
23. Buysse D, Reynolds C. Pharmacologic treatment. In: Lichstein KL, Morin CM, editors.
Treatment of late-life insomnia. Thousand Oaks: Sage Publications; 2000. p. 231–67.
24. Greenblatt D, Harmatz J, Shader R. Clinical pharmacokinetics of anxiolytics and hypnotics
in the elderly: therapeutic considerations. Clin Pharmacokinet. 1991;21:165–77.
25. Ozdemir V, Fourie J, Busto U, Naranjo A. Pharmacokinetic changes in the elderly. Clin
Pharmacokinet. 1996;31:372–85.
26. Feely J, Coakley D. Altered pharmacodynamics in the elderly. Clin Geriatr Med.
1990;6:269–83.
27. Turner N, Scarpace PJ, Lowenthal DT. Geriatric pharmacology: basic and clinical consider-
ation. Annu Rev Pharmacol Toxicol. 1992;32:271–302.
28. Foy A, O’Connell D, Henry D, Kelly J, Cocking S, Halliday J. Benzodiazepine use
as a cause of cognitive impairment in elderly hospital inpatients. J Gerontol Med Sci.
1995;50A:M99–M106.
29. Herings R, Strieker B, de Boer A, Bakker A, Sturmans F. Benzodiazepines and the risk of
falling leading to femur fractures. Arch Intern Med. 1995;155:1801–7.
30. Swift C, Swift M, Hamley J, Stevenson I, Crooks J. Side-effect ‘tolerance’ in elderly long-
term recipients of benzodiazepine hypnotics. Age Ageing. 1987;13:335–43.
31. Petursson H, Lader M. Benzodiazepine dependence. Br J Addiction. 1984;76:133–45.
32. Schneider-Helmert D. Why low dose benzodiazepine-dependent insomniacs can’t escape
their sleeping pills. Acta Psychiatr Scand. 1988;78:706–11.
33. Grad R. Benzodiazepines for insomnia in community dwelling elderly: a review of benefit
and risk. J Fam Pract. 1995;41:473–81.
34. Wheatley D. Effects of drugs on sleep. In: Wheatley D, editor. Psychopharmacology of sleep.
New York: Raven Press; 1981. p. 153–76.
35. Tyrer P. Choices of treatment in anxiety. In: Tyrer P, editor. Psychopharmacology of anxiety.
Oxford: Oxford Medical Publications; 1989. p. 255–82.
36. Olivier H, Fitz-Gerald MJ, Babiak B. Benzodiazepines revisited. J La State Med Soc.
1988;150:483–5.
37. Brodie M. Status epilepticus in adults. BMJ. 1990;336:551–2.
38. Herrington R. The use of benzodiazepines in neuropsychiatry. In: Hindmarch I, Bauamont G,
Brandon S, Leonard B, editors. Benzodiazepines: current concepts. Chichester: John Wiley
& Sons Ltd; 1990. p. 95–110.
39. Buysse D. Drugs affecting sleep, sleepiness and performance. In: Monk TH, editor. Sleep,
sleepiness and performance. London: John Wiley & Sons; 1991. p. 249–306.
40. Morgan K. Hypnotics in the elderly: what cause for concern? Drugs. 1990;40:688–96.
41. Mangoni A, Jackson S. Age-related changes in pharmacokinetics and pharmacodynamics:
basic principles and practical applications. Br J Clin Pharmacol. 2004;57:6–14.
42. Greenblatt D, Divoll HJ, MacLaughlin D, Shader R. Kinetics and clinical effects of fluraz-
epam in young and elderly noninsomniacs. Clin Pharmacol Ther. 1981;30:475–86.
43. Swift C, Ewen J, Clarke P, Stevenson I. Responsiveness to oral diazepam in the elderly:
relationship to total and free plasma concentrations. Br J Clin Pharmacol. 1985;20:111–8.
44. Berry S, Lee Y, Cai S, Dore D. Nonbenzodiazepine sleep medication use and hip fractures in
nursing home residents. JAMA Intern Med. 2013;173:754–61.
45. Leipzig R, Cumming R, Tinetti M. Drugs and falls in older people: a systematic review and
meta-analysis: I. Psychotropic drugs. J Am Geriatr Soc [Proceedings Paper]. 1999;47:30–9.
46. Woolcott J, Richardson K, Wiens M, Patel B, Marin J, Khan K, Marra C. Meta-analysis
of the impact of 9 medication classes on falls in elderly persons. Arch Intern Med.
2009;169:1952–60.
47. Sterke C, Verhagen A, van Beeck E, van der Cammen T. The influence of drug use on
fall incidents among nursing home residents: a systematic review. Int Psychogeriatr.
2008;20:890–910.
48. Lucki I, Rickels K, Geller A. Chronic use of benzodiazepines and psychomotor and cognitive
test performance. Psychopharmacol. 1986;88:426–33.
402 M. Petrovic
71. Holton A, Riley P, Tyrer P. Factors predicting long-term outcome after chronic benzodiaz-
epine therapy. J Affect Disord. 1992;24:245–52.
72. Roy-Byme P, Hommer D. Benzodiazepine withdrawal: overview and implications for the
treatment of anxiety. Am J Med. 1988;84:1041–52.
73. Ancoli-Israel S. Sleep problems in older adults: putting myths to bed. Geriatrics.
1997;52:20–30.
74. Schweizer F, Case W, Rickels K. Benzodiazepine dependence and withdrawal in elderly
patients. Am J Psychiatry. 1989;146:529–31.
75. Petrovic M, Pevemagie D, Van Den Noortgate N, Mariman A, Michielsen W, Afschrift M. A
programme for short-term withdrawal from benzodiazepines in geriatric hospital inpatients:
success rate and effect on sleep quality. Int J Geriatr Psychiatry. 1999;14:754–60.
76. Reeve E, Ong M, Wu A, Jansen J, Petrovic M, Gnjidic D. A systematic review of interven-
tions to deprescribe benzodiazepines and other hypnotics among older people. Eur J Clin
Pharmacol. 2017;73:927–35.
77. Lader M, Tylee A, Donoghue J. Withdrawing benzodiazepines in primary care. CNS Drugs.
2009;23:19–34.
78. Morin C, Bastien C, Guay B, Radouco-Thomas M, Leblanc J, Vallières A. Randomized clini-
cal trial of supervised tapering and cognitive behavior therapy to facilitate benzodiazepine
discontinuation in older adults with chronic insomnia. Am J Psychiatry. 2004;161:332–42.
79. Lichstein K, Nau S, Wilson N, Aguillard R, Lester K, Bush A, McCrae C. Psychological
treatment of hypnotic-dependent insomnia in a primarily older adult sample. Behav Res Ther.
2013;51:787–96.
80. Gould R, Coulson M, Patel N, Highton-Williamson E, Howard R. Interventions for reduc-
ing benzodiazepine use in older people: meta-analysis of randomised controlled trials. Br J
Psychiatry. 2014;204:98–107.
81. Roberts MS, Stokes JA, King MA, Lynne T, Purdie D, Glasziou P, Wilson D, McCarthy S,
Brooks G, de Looze F, Del Mar C. Outcomes of a randomized controlled trial of a clinical
pharmacy intervention in 52 nursing homes. Br J Clin Pharmacol. 2001;51:257–65.
82. Forsetlund L, Eike MC, Gjerberg E, Vist G. Effect of interventions to reduce potentially inap-
propriate use of drugs in nursing homes: a systematic review of randomised controlled trials.
BMC Geriatr. 2011;11:16.
83. Gilbert A, Owen N, Innes J, Sansom L. Trial of an intervention to reduce chronic benzodi-
azepine use among residents of aged-care accommodation. Aust NZ J Med. 1993;23:343–7.
84. Smith A, Tett S. Improving the use of benzodiazepines—is it possible? A non-systematic
review of interventions tried in the last 20 years. BMC Health Serv Res. 2010;10:321.
85. Salzman C, Fisher J, Nobel K, Glassman R, Wolfson A, Kelley M. Cognitive improvement
following benzodiazepine discontinuation in elderly nursing home residents. Int J Geriatr
Psych. 1992;7:89–93.
86. Habraken H, Soenen K, Blondeel L, Van Elsen J, Bourda J, Coppens E, Willeput M. Gradual
withdrawal from benzodiazepines in residents of homes for the elderly: experience and sug-
gestions for future research. Eur J Clin Pharmacol. 1997;51:355–8.
87. Parr J, Kavanagh D, Young R, McCafferty K. Views of general practitioners and benzodi-
azepine users on benzodiazepines: a qualitative analysis. Soc Sci Med. 2006;62:1237–49.
88. Dyas J, Apekey T, Tilling M, Ørner R, Middleton H, Siriwardena N. Patients’ and clinicians’
experiences of consultations in primary care for sleep problems and insomnia: a focus group
study. Br J Gen Pract. 2010;60:e180–200.
89. Walsh J, Erman M, Erwin C, Jamieson A, Mahowald M, Regestein Q, Scharf M, Tigel P,
Vogel G, Ware C. Subjective hypnotic efficacy of trazodone and zolpidem in DSMIII-R pri-
mary insomnia. Hum Psychopharmacol. 1998;13:191–8.
90. Mendelson W. A review of the evidence for the efficacy and safety of trazodone in insomnia.
J Clin Psychiatry. 2005;66:469–76.
91. McCleery J, Sharpley A. Pharmacotherapies for sleep disturbances in dementia. Cochrane
Database Syst Rev. 2020;11:CD009178.
404 M. Petrovic
Age, CKD, Diabetes, Sepsis, Volume Intrinsic nephrotoxic potential, Dosage, Avoid specific drug combinations
depletion, Sodium depletion, Multiple Frequency of administrations, Duration NSAIDs and ACEI/ARB (and/or
myeloma, Acid-base disturbances, of treatment, Timing of administration, diuretics)
Hypoalbuminemia, Polypharmacy. Route of administration, Pharmaceutical Cephalosporins and
Careful assessment of: formulation. aminoglycosides
Age-related diseases (renovascular Careful assessment of: Vancomycin and aminoglycosides
disease, heart failure) Nephrotoxic potential
Cephalosporins and acyclovir
CKD, diabetes and sepsis Dose-dependent effect (mainly for
(increased risk of nephrotoxic AKI) drugs causing crystal deposition,
Volume depletion (NSAID-induced tubular toxicity, and hemodynamic
and ACEI/ARB-induced toxicity)
nephrotoxicity) Frequency of administrations
Volume and sodium depletion (aminoglycosides)
(diuretics-induced nephrotoxicity) Route of administration (intra-
Hypoalbuminemia (cisplatin- and arterial administration of contrast)
aminoglycosides-induced Pharmaceutical formulation
nephrotoxicity) (nephrotoxicity is lower with
Polypharmacy (exposure to multiple liposomal amphotericin than for
nephrotoxic drugs) amphotericin lipid complex)
Fig. 28.1 Factors to be considered for preventing nephrotoxic AKI among older patients
CKD usually coexists with other chronic diseases due to a pathogenetic link, e.g.,
diabetes and atherosclerosis as the cause of CKD, and to the sharing of risk factors
(e.g., age and smoking habits) and pathophysiological pathways described above.
Recurrent and highly prevalent clusters of diseases including CKD are the rule
according to the multimorbidity approach, which defines the main associations
between and among diseases without any hierarchical assumption. According to
such approach, Formiga et al. [7] reported that CKD contributes to a multimorbidity
cluster including coronary artery disease, hypertension, stroke, and diabetes.
Zemedikun et al. [8] showed that the multimorbidity cluster including CKD may be
wider than that previously observed, being composed by 26 conditions, among
which cardiovascular, liver, neurologic, psychiatric, respiratory, endocrine, and
muscle-skeletal diseases. More recently, cluster analysis showed that CKD may
cluster with hypertension and sensory impairments, but physical performance as
assessed by SPPB may be associated with not negligible changes in multimorbidity
clusters. These findings strengthen the need of assessing physical performance and
investigating interventions targeting physical function among complex multimorbid
patients in an attempt to improve outcomes and reduce costs associated with multi-
morbidity. Finally, CKD severity may significantly affect patterns of multimorbid-
ity, which should be also taken into account in clinical approach to multimorbid
patients [9]. In such a complex scenario, the clinical presentation of CKD may be
concealed by coexistent diseases. Thus, fatigue, sarcopenia, dyspnea, and anorexia
may be expression of several conditions. As a consequence, CKD may remain
unrecognized until blood exams performed for unrelated reasons allow detect it.
Furthermore, multimorbidity accounts for polypharmacy which, in turn, is fre-
quently nephrotoxic. The kinetics of many drugs is variously compromised in CKD,
and recognizing CKD is mandatory to tailor the doses to actual eGFR values. Thus,
renal function should systematically be screened for in the aged and multimorbid
patients. Furthermore, sarcopenia and hypoalbuminemia may decrease the distribu-
tion volume and the bound fraction of water-soluble drugs and reduce serum creati-
nine out of proportion to the true eGFR, which defines the condition of concealed
renal failure, which may further increase the risk of ADRs [10]. Indeed, diabetes
mellitus, musculoskeletal diseases, and COPD are commonly associated with this
condition [11]. Not unfrequently, acute kidney injury due to contrast medium unrav-
els CKD [12–14]. Indeed, the inherent risk starts increasing from eGFR <60 mL/
min/1.73 m2 and further increases in patients given metformin or other potentially
nephrotoxic drugs as well as in those having anemia, diabetes mellitus, CHF, and
hypotension, which are well-known age-related diseases [15].
408 A. Corsonello et al.
28.3.1 Hypertension
• Target BP levels should be 130/70 mmHg (and not below) for older people of
65 years or more; in older patients over 80 years or in all frail older patients, a
smaller reduction of SBP is advised according to the benefit/risk ratio of treat-
ment. These target thresholds are higher compared with those suggested from
clinical practice guidelines [20] and are derived from independent studies show-
ing how a more intensive BP treatment may worsen prognosis in such popula-
tions [21]. Diastolic blood pressure levels <70 mmHg were in fact associated
with increased cardiovascular risk [21] and accelerated decline in kidney func-
tion in patients aged 85 years or more [22]; similarly, systolic blood pressure
<130 mmHg was associated with increased mortality in CKD patients aged
75 years or more [23].
• Angiotensin-converting enzyme inhibitors (ACE-I) and angiotensin II receptor
antagonists (ARBs) are still considered first-line agents for patients with diabetic
CKD (defined as an eGFR <60 mL/min per 1.73 m2 or micro- or macroalbumin-
uria) and nondiabetic proteinuric CKD [24], even in the absence of hypertension
[25, 26]. Usage of renin angiotensin system (RAS) blockers is important to slow
progression of CKD, thanks to their antiproteinuric effects. However, potential
iatrogenic risks related to ACE-I or ARBs should be taken into account. Indeed,
prescription of ACE-I and ARBs in older patients with CKD mandates careful
monitoring for acute renal failure and hyperkalemia; the occurrence of symp-
tomatic hypotension or eGFR <15 mL/min/1.73 m2 suggests discontinuation of
treatment, whereas reduction of drug doses is indicated for milder renal function
28 Optimizing Pharmacotherapy in Older Patients: An Interdisciplinary Approach… 409
28.3.2 Diabetes
kidney function may result in the accumulation of several drugs and/or their
metabolites.
[30]. Conversely, DPP-4 inhibitors are potentially advantageous for older dia-
betic patients with CKD because they are associated with a lower risk of hypo-
glycemia compared to other antidiabetic medications [30]. All currently available
DPP-4 inhibitors can be used in CKD, but dosage of sitagliptin, saxagliptin, and
alogliptin needs to be reduced in patients with GFR <50 mL/min/1.73 m2 [30,
36]. Linagliptin is not cleared by the kidney and does not require any dosage
adjustment in patients with CKD [30, 37].
–– Inhibitors of sodium-glucose cotransporters type 2 (SGLT2) significantly and
consistently improve glycemic control with a marginal risk of hypoglycemia,
especially when in monotherapy [30]. Furthermore, they have recently
emerged as a disease-modifying treatment to counteract the progression of
chronic renal disease in patients with or without diabetes [38]. No dose adjust-
ment is required in mild CKD, but current evidence suggests being very cau-
tious in moderate-severe stages of the disease. Indeed, findings from
DECLARE-TIMI 58 [39, 40], CREDENCE [41], DAPA-CKD [42], and
EMPA-KIDNEY trials have shown that these drugs can be safely prescribed
after dose adjustment in patients with eGFR ≥20–30 mL/min/1.73 m2; con-
versely, their use in patients with lower eGFR is not recommended. Within
this family, canagliflozin has good safety profile in older diabetic patients
with macroalbuminuria [43], whereas dapagliflozin can be safely prescribed
to decrease the risk of CKD progression irrespective of albuminuria [44];
previous evidence of an increased risk of lower limb amputations in patients
taking canagliflozin [35] has not been confirmed by recent RCTs, while con-
cerns of genital mycotic infections and euglycemic diabetic ketoacidosis were
confirmed for canagliflozin only [41, 42].
• Insulin remains the mainstay of treatment in diabetic patients with secondary
failure and moderate to advanced CKD, especially those undergoing dialysis
[30]. About one third of exogenous insulin is cleared by the kidney. Sixty percent
of the renal clearance is due to glomerular filtration, while the remaining 40% is
secreted after uptake from peritubular vessels. The available long- and short-
acting insulin analogues have similar pharmacokinetics in patients with CKD
[30]. To reduce the risk of hypoglycemia, insulin dosage reduction of 25% has
been recommended in patients with GFR = 10–50 mL/min. However, when the
long-acting insulins glargine or detemir are used, it may be wise to start treat-
ment with 50% of the usual initial dosage of 0.1 IU/kg, titrating the dosage until
target fasting glucose concentrations are reached [45]. The pharmacokinetics of
the ultralong-acting insulin degludec does not change in patients with different
degrees of kidney dysfunction [32, 46], thus suggesting that dose adjustments in
patients with mild to severe renal impairment should not be required.
28.3.3 Albuminuria
the progression of diabetic kidney disease and improve clinical outcomes even in
the absence of hypertension [47]. Guidelines recommend that normotensive people
with diabetes and macroalbuminuria should be treated with ACE-I or ARBs [19].
However, treatment with ACE-I or ARBs was found beneficial even in microalbu-
minuric normotensive patients, where benefits were more pronounced in diabetic
compared to nondiabetic patients and in patients with more severe proteinuria
(>3 g/24 h) compared to those with less severe proteinuria (<1 g/24 h) [19, 48].
Evidence from subgroup analysis of older patients from proteinuria-lowering trials
suggests a reduced risk of cardiovascular outcomes even in this group of patients
compared with that seen in younger subjects [49].
However, concerns related to treatment outcomes in older patients already
described above (see paragraph dealing with hypertension) also apply to the use of
these medications in normotensive patients. Additionally, combined therapy with
ACE-I and ARBs was proposed to treat proteinuria, but the ONTARGET study
clearly showed that combining ACE-I and ARBs was associated with faster decline
in renal function, more side effects, and worse overall outcome [50, 51]. These find-
ings are of particular concerns for older complex patients who are much more vul-
nerable to iatrogenic injury.
28.3.4 Hyperlipidemia
Lipid metabolism abnormalities are commonly associated with CKD and contribute
to cardiovascular morbidity and mortality in CKD patients.
patients with persistently high PTH levels (>2.3–3 times the normal values),
despite treatment of hyperphosphatemia and vitamin D deficiency, calcitriol
should be administered with a regimen of 0.25 mcg three times a week [64].
• Hypercalcemia: in CKD stages 3 to 5, total elemental calcium intake including
dietary and supplements should not exceed 2000 mg/d. The National Osteoporosis
Foundation recommends a daily calcium intake of 1200 mg and vitamin D intake
of 800–1000 IU/d for adults 50 and older [64, 68]. However, an only modestly
increased risk of cardiovascular events in postmenopausal women undergoing
calcium supplementation with or without vitamin D was reported in some stud-
ies [69, 70], thus suggesting the need to weigh the clinical significance of cardio-
vascular risks related to calcium supplements against the risks related to
osteoporosis and its complications.
• Osteoporosis in CKD patients:
–– Use of bisphosphonates in CKD older patients with osteoporosis is controver-
sial, as it was found associated with progressive renal disease, acute renal
failure, and nephrotic syndrome [71]. This finding was not confirmed in other
studies showing that even in older, frail, osteoporotic patients with renal
impairment, intravenous bisphosphonate therapy did not result in long-term
renal function decline [72]. However, it is worth reminding that bisphospho-
nates are not indicated in adynamic or osteomalacic bone disease. Additionally,
calcium-phosphorous alterations, vitamin D deficiency, and hyperparathy-
roidism should always be addressed before starting bisphosphonates. Finally,
there is no evidence that using bisphosphonates for short periods of time
(2–3 years) in the CKD population will result in a significantly reduced inci-
dence of fractures [71, 73].
–– The anti-RANKL monoclonal antibody denosumab decreases bone reabsorp-
tion, is useful for osteoporosis treatment and prevention of fractures, and is
not cleared by the kidney. The efficacy and renal safety of denosumab have
been demonstrated in elderly women [74]. However, by inhibiting bone
resorption, it may lead to hypocalcemia. For this reason, patients with creati-
nine clearance <30 mL/min and/or other conditions predisposing to hypocal-
cemia should be strictly monitored during denosumab treatment [75, 76].
–– The use of selective estrogen receptor modulators (SERM) raloxifene was
found associated with increased spine BMD, reduced incidence of vertebral
fractures, and no effect on non-vertebral fractures compared with placebo in
postmenopausal women with osteoporosis. No safety risk related to raloxi-
fene in patients with mild to moderate CKD over the 2- to 3-year observation
period [64, 77]. However, SERMs may increase the risk of deep venous
thrombosis (DVT), pulmonary embolism, and fatal stroke in postmenopausal
women with coronary heart disease, which raise the need for further investi-
gations about their long-term safety in older patients already carrying
increased risk of cardiovascular events [64].
–– Strontium ranelate is able to rebalance bone remodeling without a strong inhi-
bition of bone reabsorption, which could reduce the risk of hypocalcemia
during treatment. However, it is cleared by the kidney and is contraindicated
in patients with GFR <30 mL/min. Additionally, an increased cardiovascular
28 Optimizing Pharmacotherapy in Older Patients: An Interdisciplinary Approach… 415
risk was observed in a pooled analysis of strontium RCTs [78, 79], which
makes this drug less appealing for use in older patients with CKD.
28.3.6 Acidosis
• Start oral alkali therapy with sodium bicarbonate (or citrate) when serum bicar-
bonate <22 mmol/L [81].
• Therapy should be given to maintain the normal range (23–29 mmol/L) [82],
since small clinical trials have reported that treatment within this range may
delay CKD progression [83, 84], improve nutrition status [85], increase insulin
sensitivity [86], and decrease atherogenic lipid levels [87]. Conversely, the effect
on bone health was not fully documented [88]. However, especially in older
patients, overcorrection should be avoided because of potentially harmful effects,
mainly resulting from the U-shaped relationship between serum bicarbonate lev-
els and mortality [89]. Whether the sodium load that accompanies alkali therapy
could cause volume overload and hypertension remains to be determined.
Additionally, alkali therapy may lead to hypokalemia, ionized hypocalcemia,
and QTc prolongation. The above evidence supports a judicious use of alkali
treatment as well as a careful monitoring of serum bicarbonates, electrolytes, and
ECG in older patients with acidosis.
28.3.7 Anemia
The association between CKD and anemia is related to several mechanisms such as
impaired production of erythropoietin, iron deficiency, and inflammation with
increased hepcidin levels [90]. Despite its prevalence increases with decline of
eGFR, in some older patients, e.g., those with diabetic kidney disease, impaired
production of erythropoietin usually precedes the decline of eGFR, and even mildly
depressed eGFR may then be associated with significant anemia of renal origin
[91]. Recommendations for treatment of CKD-related anemia are mainly based on
clinical practice guidelines, although most studies did not include geriatric popula-
tions [92]:
%HRC and CHr are not available or the patient has thalassaemia/thalassaemia
trait, check transferrin saturation (TSAT), and ferritin serum levels.
• Start iron supplementation before erythropoiesis stimulating factors (ESAs) in
patients with iron responsiveness, defined as having %HRC >6% and CHR
>29 pg (or TSAT <20% and ferritin <100 μg/L). Oral iron (ferrous sulfate, fuma-
rate, or gluconate) is the preferred starting treatment, at a daily regimen of
200 mg in up to three divided doses. Target iron status is defined as a %HRC
<6% (unless ferritin >800 μg/L) and CHr >29 pg (or TSAT >20% and ferritin
>100 μg/L) when above tests are not available or patient has thalassaemia/thal-
assaemia trait; serum ferritin levels should be checked every 1–3 months to
check for potential iron overload.
• Start ESA for patients who are likely to benefit in quality of life and physical
function, only after correcting iron status (patients with %HRC >6% and CHr
>29 pg). Although patients of 75 years or more were excluded from studies
assessing ESA efficacy in the treatment of CKD-related anemia [90], guidelines
clearly suggest that age alone should not be a determinant for treating or not
treating anemia in CKD [90]. However, long-term treatment with ESA has been
associated with increased blood pressure and seizures [94], as well as polyglobu-
lia and increased thrombotic risk. For this reason, older patients should always
be closely monitored, and ESAs should be given at the lowest dose capable of (a)
maintaining stable Hb between 10.0 and 12.0 g/dL and (b) keeping rate of Hb
increase between 1.0 and 2.0 g/dL/month. Caution should be taken in some cat-
egories, including patients with previous cancer or previous/current stroke (risk
of recurrence) and older patients with increased thrombotic risk, bedridden
patients, and individuals with very limited functional capacity (in whom risks
clearly outweigh the benefits of treatment).
Among older CKD patients, ESA administration may be not sufficient to coun-
teract anemia due to the coexistence of systemic inflammatory processes blunting
response to erythropoietin, and inflammation-induced increased hepcidin produc-
tion reducing iron absorption and utilization [90], which further strengthen the need
for studies in older patients with multimorbidity.
28.4 Nephrotoxicity
Medications can impair any of the kidney functions, including renal arterial blood
flow, glomerular filtration, tubular fluid formation, and urine output. While many
drugs have a single mechanism of injury, some medications may affect kidney func-
tion by multiple pathways. Main mechanisms of nephrotoxicity are summarized in
Table 28.1 [95, 96].
Overall, nephrotoxic medications contribute to up to 66% of all acute kidney
injuries (AKIs) in older hospitalized patients [97]. Older age is an independent pre-
dictor of AKI in several different study populations [98–105]. In particular, the rela-
tive risk of developing AKI after myocardial revascularization with or without
28 Optimizing Pharmacotherapy in Older Patients: An Interdisciplinary Approach… 417
concurrent valvular surgery increases from 1.6 (95% CI, 1.1–2.3) among patients
aged 70–79 years to 3.5 (95%CI, 1.9–6.3) among patients aged 80–95 years com-
pared to those aged less than 70 [106]. The risk of developing AKI was 50% higher
in patients admitted to intensive care units aged 65 or more compared to younger
ones [102]. Nevertheless, only few studies were devoted to address nephrotoxic
AKI among older patients. In a single-center Japanese study of hospitalized patients
aged 80 or more, the use of nephrotoxic medications (with antibiotics being the
most common culprit medications) was among major causes of AKI, together with
hypovolemia, cardiac dysfunction, and respiratory failure [107]. In other studies,
the relationship between age and risk of nephrotoxic AKI was no longer significant
after adjusting for confounders [108, 109]. However, it should be considered that
older patients are frequently affected by multiple chronic diseases which need to be
treated with complex polypharmacy regimens. In this scenario, both multimorbidity
per se and the use of multiple nephrotoxic medications may contribute to increase
the risk of AKI [96, 108–110]. Older age is also associated with increased risk of
nephrotoxic AKI-related death [96]. More recently, glycopeptide antibiotics, drugs
418 A. Corsonello et al.
Preventing AKI first involves recognizing the increased vulnerability of the aging
kidney to injury due to age-related structural and functional changes. Indeed, age
>75 years is a recognized risk factor for nephrotoxicity [110]. However, vulnerabil-
ity to nephrotoxic injury may be further increased by multiple chronic diseases
accumulated over the life span, and especially CKD. Thus, identifying CKD patients
may help to individuate patients at greater risk of nephrotoxicity. Other diseases
associated with increased risk of developing AKI are diabetes, heart failure, liver
disease, peripheral arterial disease, hypotension, hypovolemia, and acid-base disor-
ders [116]. Physicians should be aware of such an increased risk when prescribing
a potentially nephrotoxic medication for patients carrying one or more of these
conditions.
General rules to reduce the risk of nephrotoxic AKI include assessing baseline
renal function and consider patient’s renal function when prescribing a new drug,
adjusting medication dosages to renal function, avoiding dangerous nephrotoxic
combinations (e.g., the triple whammy, cephalosporins plus aminoglycosides, van-
comycin plus aminoglycosides, cephalosporins plus acyclovir), and correcting,
when possible, risk factors for nephrotoxicity before the initiation of drug therapy.
Adequate attention should be also devoted to ensure valid hydration before and dur-
ing therapy with potential nephrotoxins, use equally effective non-nephrotoxic
drugs whenever possible, monitoring drug levels to use the correct dose [117].
Improving overall quality of prescriptions may also contribute to reducing risks. In
a study of hospitalized patients with a minimum 0.5 mg/dL change in serum creati-
nine who were prescribed a nephrotoxic or renal eliminated medication, failure to
adjust dosing for kidney function (63%) and the use of nephrotoxic medications
during AKI accounted for 63% and 28% of all recorded adverse drug events [118].
Thus, reducing inappropriate prescribing is very likely to represent a good strategy
for reducing the risk of nephrotoxicity, and studies using available explicit criteria
to target inappropriate medications in older patients [119, 120] are highly desirable.
A summary of useful strategies is suggested in Fig. 28.1.
Specific recommendations exist for preventing radiocontrast-induced AKI
(CI-AKI), which is among the most common cause of hospital-acquired AKI. Prior
28 Optimizing Pharmacotherapy in Older Patients: An Interdisciplinary Approach… 419
to contrast media exposure, serum creatinine must be measured and GFR calcu-
lated. Patients with GFR less than 60 mL/min/1.73 m2 should be considered at risk.
Volume expansion with either isotonic sodium chloride or sodium bicarbonate solu-
tions may be useful to prevent CI-AKI in patients with reduced volemia. The use of
established nephrotoxic drugs (e.g., cyclosporine, aminoglycoside, NSAIDs,
COX-2 inhibitors) should be stopped for at least 2 days. Diabetic patients with pre-
existing renal impairment should withhold metformin for 48 h because lactic acido-
sis may occur once CI-AKI develops [121]. Finally, GFR should always be estimated
48–72 h after receiving contrast media in order to ensure timely CI-AKI detection.
Preventing strategies include isotonic fluid administration, N-acetylcysteine,
alkalization of the urine with IV or oral agents, and statins. Volume expansion with
either isotonic sodium chloride or sodium bicarbonate solutions may be useful to
prevent CI-AKI in patients with reduced volemia. While no specific benefits have
been observed when using sodium bicarbonate vs. isotonic saline likely because
target urine alkalization was not always reached [122, 123], the combination of oral
sodium/potassium citrate and volume expansion with isotonic saline was found to
significantly reduce the risk of CI-AKI [124]. Volume expansion often raises the
concern related to the risk of fluid overload in patients with impaired renal function
or heart failure. Using left ventricular end-diastolic pressure (LVEDP) to guide vol-
ume expansion was found to reduce the risk of CI-AKI but not that of stopping fluid
administration because of the occurrence of dyspnea [125]. The Prevention of
Radiocontrast Induced Nephropathy Clinical Evaluation (PRINCE) trial showed
that forcing diuresis with loop diuretics with urinary flow rate >150 mL/h may pro-
tect against CI-AKI, even if the risk of volume depletion needs to be taken into
account [126]. However, forced diuresis in combination with saline infusion to
match the input and maintain iso-volemia was found to lower the risk of
CI-AKI [127].
N-acetylcysteine did not show any significant benefit in terms of reduction of
CI-AKI risk in both low-risk or high-risk populations [128, 129], while rosuvastatin
was found to reduce the likelihood of CI-AKI [130, 131].
28.5 Conclusions
References
1. KDIGO CKD Work Group. KDIGO 2012 clinical practice guideline for the evaluation and
management of chronic kidney disease. Kidney Int Suppl. 2013;3:1–150.
2. Collins AJ, Foley RN, Chavers B, Gilbertson D, Herzog C, Johansen K, et al. United States
Renal Data System 2011 Annual Data Report: Atlas of chronic kidney disease & end-stage
renal disease in the United States. Am J Kidney Dis. 2012;59(1 Suppl 1):A7, e1–420.
3. Aucella F, Corsonello A, Leosco D, Brunori G, Gesualdo L, Antonelli-Incalzi R. Beyond
chronic kidney disease: the diagnosis of Renal Disease in the Elderly as an unmet need.
A position paper endorsed by Italian Society of Nephrology (SIN) and Italian Society of
Geriatrics and Gerontology (SIGG). J Nephrol. 2019;32(2):165–76.
4. Webster AC, Nagler EV, Morton RL, Masson P. Chronic kidney disease. Lancet.
2017;389(10075):1238–52.
5. Glassock RJ. Con: Thresholds to define chronic kidney disease should not be age dependent.
Nephrol Dial Transplant. 2014;29(4):774–9; discussion 9–82.
6. Glassock R, Delanaye P, El Nahas M. An age-calibrated classification of chronic kidney
disease. JAMA. 2015;314(6):559–60.
7. Formiga F, Ferrer A, Sanz H, Marengoni A, Alburquerque J, Pujol R, et al. Patterns of
comorbidity and multimorbidity in the oldest old: the Octabaix study. Eur J Intern Med.
2013;24(1):40–4.
8. Zemedikun DT, Gray LJ, Khunti K, Davies MJ, Dhalwani NN. Patterns of multimorbid-
ity in middle-aged and older adults: an analysis of the UK Biobank data. Mayo Clin Proc.
2018;93(7):857–66.
9. Corsonello A, Fabbietti P, Formiga F, Moreno-Gonzalez R, Tap L, Mattace-Raso F, et al.
Chronic kidney disease in the context of multimorbidity patterns: the role of physical perfor-
mance : The screening for CKD among older people across Europe (SCOPE) study. BMC
Geriatr. 2020;20(Suppl 1):350.
10. Corsonello A, Pedone C, Corica F, Mussi C, Carbonin P, Antonelli Incalzi R, et al. Concealed
renal insufficiency and adverse drug reactions in elderly hospitalized patients. Arch Intern
Med. 2005;165(7):790–5.
11. Incalzi RA, Corsonello A, Pedone C, Battaglia S, Paglino G, Bellia V, et al. Chronic renal
failure: a neglected comorbidity of COPD. Chest. 2010;137(4):831–7.
12. Chawla LS, Amdur RL, Amodeo S, Kimmel PL, Palant CE. The severity of acute kidney
injury predicts progression to chronic kidney disease. Kidney Int. 2011;79(12):1361–9.
13. Ishani A, Xue JL, Himmelfarb J, Eggers PW, Kimmel PL, Molitoris BA, et al. Acute kidney
injury increases risk of ESRD among elderly. J Am Soc Nephrol. 2009;20(1):223–8.
14. Bucaloiu ID, Kirchner HL, Norfolk ER, Hartle JE 2nd, Perkins RM. Increased risk of death
and de novo chronic kidney disease following reversible acute kidney injury. Kidney Int.
2012;81(5):477–85.
15. Mehran R, Aymong ED, Nikolsky E, Lasic Z, Iakovou I, Fahy M, et al. A simple risk score for
prediction of contrast-induced nephropathy after percutaneous coronary intervention: devel-
opment and initial validation. J Am Coll Cardiol. 2004;44(7):1393–9.
16. Ku E, Lee BJ, Wei J, Weir MR. Hypertension in CKD: core curriculum 2019. Am J Kidney
Dis. 2019;74(1):120–31.
17. Bakris G, Ali W, Parati G. ACC/AHA versus ESC/ESH on hypertension guidelines: JACC
guideline comparison. J Am Coll Cardiol. 2019;73(23):3018–26.
18. Verbeke F, Lindley E, Van Bortel L, Vanholder R, London G, Cochat P, et al. A European
Renal Best Practice (ERBP) position statement on the Kidney Disease: Improving Global
Outcomes (KDIGO) clinical practice guideline for the management of blood pressure in non-
dialysis-dependent chronic kidney disease: an endorsement with some caveats for real-life
application. Nephrol Dial Transplant. 2014;29(3):490–6.
19. Cheung A, Chang TI, Cushman W, Furth S, Hou FF, Ix JH, et al. KDIGO (Kidney Disease:
Improving Global Outcomes) guideline update on the management of blood pressure in
28 Optimizing Pharmacotherapy in Older Patients: An Interdisciplinary Approach… 421
chronic kidney disease: what’s new and what’s different from other guidelines. Hypertension.
2021;78(suppl_1):A52. -A.
20. Williams B, Mancia G, Spiering W, Agabiti Rosei E, Azizi M, Burnier M, et al. ESC/ESH
Guidelines for the management of arterial hypertension: the task force for the management of
arterial hypertension of the European Society of Cardiology (ESC) and the European Society
of Hypertension (ESH). Eur Heart J. 2018;39(33):3021–104.
21. Franklin SS, Gokhale SS, Chow VH, Larson MG, Levy D, Vasan RS, et al. Does low diastolic
blood pressure contribute to the risk of recurrent hypertensive cardiovascular disease events?
Hypertension. 2015;65(2):299–305.
22. van Bemmel T, Woittiez K, Blauw GJ, van der Sman-de BF, Dekker FW, Westendorp RG,
et al. Prospective study of the effect of blood pressure on renal function in old age: the Leiden
85-Plus Study. J Am Soc Nephrol. 2006;17(9):2561–6.
23. Weiss JW, Johnson ES, Petrik A, Smith DH, Yang X, Thorp ML. Systolic blood pressure
and mortality among older community-dwelling adults with CKD. Am J Kidney Dis.
2010;56(6):1062–71.
24. Cheung AK, Chang TI, Cushman WC, Furth SL, Hou FF, Ix JH, et al. KDIGO 2021 clini-
cal practice guideline for the management of blood pressure in chronic kidney disease.
2021;99(3):S1–S87.
25. Taler SJ, Agarwal R, Bakris GL, Flynn JT, Nilsson PM, Rahman M, et al. KDOQI US com-
mentary on the 2012 KDIGO clinical practice guideline for management of blood pressure in
CKD. Am J Kidney Dis. 2013;62(2):201–13.
26. Page MR, The JNC. 8 hypertension guidelines: an in-depth guide. Am J Manag Care.
2014;20(1):E8.
27. Agarwal R, Sinha AD, Cramer AE, Balmes-Fenwick M, Dickinson JH, Ouyang F, et al.
Chlorthalidone for hypertension in advanced chronic kidney disease. New Engl J Med.
2021;385(27):2507–19.
28. Douros A, Ebert N, Jakob O, Martus P, Kreutz R, Schaeffner E. Estimating kidney function
and use of oral antidiabetic drugs in elderly. Fundam Clin Pharmacol. 2015;29(3):321–8.
29. de Boer IH, Caramori ML, Chan JCN, Heerspink HJL, Hurst C, Khunti K, et al. KDIGO
2020 clinical practice guideline for diabetes management in chronic kidney disease. Kidney
Int. 2020;98(4):S1–S115.
30. de Boer IH, Caramori ML, Chan JC, Heerspink HJ, Hurst C, Khunti K, et al. Executive sum-
mary of the 2020 KDIGO diabetes management in CKD guideline: evidence-based advances
in monitoring and treatment. Kidney Int. 98:2020, 839–848.
31. Inzucchi SE, Lipska KJ, Mayo H, Bailey CJ, McGuire DK. Metformin in patients with type 2
diabetes and kidney disease: a systematic review. JAMA. 2014;312(24):2668–75.
32. Navaneethan SD, Zoungas S, Caramori ML, Chan JC, Heerspink HJ, Hurst C, et al. Diabetes
management in chronic kidney disease: synopsis of the 2020 KDIGO clinical practice guide-
line. 2021;174(3):385–94.
33. KDOQI clinical practice guideline for diabetes and CKD: 2012 Update. Am J Kidney Dis.
2012;60(5):850–86.
34. Wang W, Zhou X, Kwong JSW, Li L, Li Y, Sun X. Efficacy and safety of thiazolidinediones
in diabetes patients with renal impairment: a systematic review and meta-analysis. Sci Rep.
2017;7(1):1717.
35. Palmer SC, Tendal B, Mustafa RA, Vandvik PO, Li S, Hao Q, et al. Sodium-glucose cotrans-
porter protein-2 (SGLT-2) inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists
for type 2 diabetes: systematic review and network meta-analysis of randomised controlled
trials. BMJ (Clinical research ed). 2021;372:m4573.
36. Aroda VR, Henry RR, Han J, Huang W, DeYoung MB, Darsow T, et al. Efficacy of GLP-1
receptor agonists and DPP-4 inhibitors: meta-analysis and systematic review. Clin Ther.
2012;34(6):1247–58 e22.
37. Giorda CB, Nada E, Tartaglino B. Pharmacokinetics, safety, and efficacy of DPP-4 inhibitors
and GLP-1 receptor agonists in patients with type 2 diabetes mellitus and renal or hepatic
impairment. A systematic review of the literature. Endocrine. 2014;46(3):406–19.
422 A. Corsonello et al.
38. Yau K, Dharia A, Alrowiyti I, Cherney DZI. Prescribing SGLT2 inhibitors in patients with
CKD: expanding indications and practical considerations. Kidney Int Rep. 2022;7(7):1463–76.
39. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, et al. Dapagliflozin and car-
diovascular outcomes in type 2 diabetes. N Engl J Med. 2019;380(4):347–57.
40. Mosenzon O, Wiviott SD, Cahn A, Rozenberg A, Yanuv I, Goodrich EL, et al. Effects of
dapagliflozin on development and progression of kidney disease in patients with type 2 diabe-
tes: an analysis from the DECLARE-TIMI 58 randomised trial. Lancet Diabetes Endocrinol.
2019;7(8):606–17.
41. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al.
Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med.
2019;380(24):2295–306.
42. Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, et al.
Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436–46.
43. Sehgal V, Bajwa SJ, Sehgal R, Consalvo JA. Management of diabetes in the elderly with
canagliflozin: a newer hypoglycemic drug on the horizon. J Pharmacol Pharmacother.
2014;5(4):227–31.
44. Mende CW. Chronic kidney disease and SGLT2 inhibitors: a review of the evolving treatment
landscape. Adv Ther. 2022;39(1):148–64.
45. Scheen AJ. Pharmacokinetic considerations for the treatment of diabetes in patients with
chronic kidney disease. Expert Opin Drug Metab Toxicol. 2013;9(5):529–50.
46. Kiss I, Arold G, Roepstorff C, Bottcher SG, Klim S, Haahr H. Insulin degludec: pharmacoki-
netics in patients with renal impairment. Clin Pharmacokinet. 2014;53(2):175–83.
47. Murton M, Goff-Leggett D, Bobrowska A, Garcia Sanchez JJ, James G, Wittbrodt E, et al.
Burden of chronic kidney disease by KDIGO categories of glomerular filtration rate and
albuminuria: a systematic review. Adv Ther. 2021;38(1):180–200.
48. Verma V, Kant R, Sunnoqrot N, Gambert SR. Proteinuria in the elderly: evaluation and man-
agement. Int Urol Nephrol. 2012;44(6):1745–51.
49. Winkelmayer WC, Zhang Z, Shahinfar S, Cooper ME, Avorn J, Brenner BM. Efficacy and
safety of angiotensin II receptor blockade in elderly patients with diabetes. Diabetes Care.
2006;29(10):2210–7.
50. Mann JF, Schmieder RE, McQueen M, Dyal L, Schumacher H, Pogue J, et al. Renal outcomes
with telmisartan, ramipril, or both, in people at high vascular risk (the ONTARGET study):
a multicentre, randomised, double-blind, controlled trial. Lancet. 2008;372(9638):547–53.
51. Ren F, Tang L, Cai Y, Yuan X, Huang W, Luo L, et al. Meta-analysis: the efficacy and
safety of combined treatment with ARB and ACEI on diabetic nephropathy. Ren Fail.
2015;37(4):548–61.
52. Armitage J, Baigent C, Barnes E, Betteridge DJ, Blackwell L, Blazing M, et al. Efficacy and
safety of statin therapy in older people: a meta-analysis of individual participant data from 28
randomised controlled trials. Lancet. 2019;393(10170):407–15.
53. Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM, Cobbe SM, et al. Pravastatin
in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial.
Lancet. 2002;360(9346):1623–30.
54. Wanner C, Krane V, Marz W, Olschewski M, Mann JF, Ruf G, et al. Atorvastatin in patients
with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med. 2005;353(3):238–48.
55. Baigent C, Landray MJ, Reith C, Emberson J, Wheeler DC, Tomson C, et al. The effects of
lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney
disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet.
2011;377(9784):2181–92.
56. Fellstrom BC, Jardine AG, Schmieder RE, Holdaas H, Bannister K, Beutler J, et al.
Rosuvastatin and cardiovascular events in patients undergoing hemodialysis. N Engl J Med.
2009;360(14):1395–407.
57. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, et al. 2018 AHA/
ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on
the management of blood cholesterol: a report of the American College of Cardiology/
28 Optimizing Pharmacotherapy in Older Patients: An Interdisciplinary Approach… 423
79. Ott SMJNRN. Therapy for patients with CKD and low bone mineral density. Nat Rev
Nephrol. 2013;9(11):681–92.
80. Sy J, Chen JLT, Kalantar-Zadeh K. New solutions to old problems-metabolic acidosis in
chronic kidney disease. Ann Transl Med. 2020;8(19):1256.
81. Andrassy KM. Comments on ‘KDIGO 2012 clinical practice guideline for the evaluation and
management of chronic kidney disease’. Kidney Int. 2013;84(3):622–3.
82. Melamed ML, Raphael KL. Metabolic acidosis in CKD: a review of recent findings. Kidney
Med. 2021;3(2):267–77.
83. Mannon EC, O’Connor PM. Alkali supplementation as a therapeutic in chronic kidney dis-
ease: what mediates protection? Am J Physiol-Renal Physiol. 2020;319(6):F1090–F104.
84. Di Iorio BR, Bellasi A, Raphael KL, Santoro D, Aucella F, Garofano L, et al. Treatment of
metabolic acidosis with sodium bicarbonate delays progression of chronic kidney disease:
the UBI Study. J Nephrol. 2019;32(6):989–1001.
85. Mathur RP, Dash SC, Gupta N, Prakash S, Saxena S, Bhowmik D. Effects of correction of
metabolic acidosis on blood urea and bone metabolism in patients with mild to moderate
chronic kidney disease: a prospective randomized single blind controlled trial. Renal Fail.
2006;28(1):1–5.
86. Bellasi A, Di Micco L, Santoro D, Marzocco S, De Simone E, Cozzolino M, et al. Correction
of metabolic acidosis improves insulin resistance in chronic kidney disease. BMC Nephrol.
2016;17(1):158.
87. Rizzetto F, Mafra D, Barra AB, Pires de Melo G, Abdalla DSP, Leite M Jr. One-year con-
servative care using sodium bicarbonate supplementation is associated with a decrease in
electronegative LDL in chronic kidney disease patients: a pilot study. Cardiorenal Med.
2017;7(4):334–41.
88. Melamed ML, Horwitz EJ, Dobre MA, Abramowitz MK, Zhang L, Lo Y, et al. Effects of
sodium bicarbonate in CKD stages 3 and 4: a randomized, placebo-controlled, multicenter
clinical trial. Am J Kidney Dis. 2020;75(2):225–34.
89. Kovesdy CP, Anderson JE, Kalantar-Zadeh K. Association of serum bicarbonate levels
with mortality in patients with non-dialysis-dependent CKD. Nephrol Dial Transplant.
2009;24(4):1232–7.
90. Portolés J, Martín L, Broseta JJ, Cases A. Anemia in chronic kidney disease: from patho-
physiology and current treatments, to future agents. Front Med (Lausanne). 2021;8(328).
[Review].
91. Bosman DR, Winkler AS, Marsden JT, Macdougall IC, Watkins PJ. Anemia with erythro-
poietin deficiency occurs early in diabetic nephropathy. Diabetes Care. 2001;24(3):495–9.
92. Ratcliffe LE, Thomas W, Glen J, Padhi S, Pordes BA, Wonderling D, et al. Diagnosis and
management of iron deficiency in CKD: a summary of the NICE guideline recommendations
and their rationale. Am J Kidney Dis. 2016;67(4):548–58.
93. National Clinical Guideline Centre (UK). National Institute for Health and Care Excellence:
clinical guidelines. anaemia management in chronic kidney disease: partial Update 2015.
London: Royal College of Physicians (UK); 2015.
94. Solomon SD, Uno H, Lewis EF, Eckardt KU, Lin J, Burdmann EA, et al. Erythropoietic
response and outcomes in kidney disease and type 2 diabetes. N Engl J Med.
2010;363(12):1146–55.
95. Nolin TD, Himmelfarb J. Mechanisms of drug-induced nephrotoxicity. Handb Exp
Pharmacol. 2010;196:111–30.
96. Fusco S, Garasto S, Corsonello A, Vena S, Mari V, Gareri P, et al. Medication-induced neph-
rotoxicity in older patients. Curr Drug Metab. 2016;17(6):608–25.
97. Kohli HS, Bhaskaran MC, Muthukumar T, Thennarasu K, Sud K, Jha V, et al. Treatment-
related acute renal failure in the elderly: a hospital-based prospective study. Nephrol Dial
Transplant. 2000;15(2):212–7.
98. Groeneveld AB, Tran DD, van der Meulen J, Nauta JJ, Thijs LG. Acute renal failure in
the medical intensive care unit: predisposing, complicating factors and outcome. Nephron.
1991;59(4):602–10.
28 Optimizing Pharmacotherapy in Older Patients: An Interdisciplinary Approach… 425
99. Zanardo G, Michielon P, Paccagnella A, Rosi P, Calo M, Salandin V, et al. Acute renal failure
in the patient undergoing cardiac operation. Prevalence, mortality rate, and main risk factors.
J Thorac Cardiovasc Surg. 1994;107(6):1489–95.
100. Vivino G, Antonelli M, Moro ML, Cottini F, Conti G, Bufi M, et al. Risk factors for acute
renal failure in trauma patients. Intensive Care Med. 1998;24(8):808–14.
101. Conlon PJ, Stafford-Smith M, White WD, Newman MF, King S, Winn MP, et al. Acute renal
failure following cardiac surgery. Nephrol Dial Transplant. 1999;14(5):1158–62.
102. de Mendonca A, Vincent JL, Suter PM, Moreno R, Dearden NM, Antonelli M, et al. Acute
renal failure in the ICU: risk factors and outcome evaluated by the SOFA score. Intensive
Care Med. 2000;26(7):915–21.
103. Bove T, Calabro MG, Landoni G, Aletti G, Marino G, Crescenzi G, et al. The incidence and
risk of acute renal failure after cardiac surgery. J Cardiothorac Vasc Anesth. 2004;18(4):442–5.
104. Bahar I, Akgul A, Ozatik MA, Vural KM, Demirbag AE, Boran M, et al. Acute renal failure
following open heart surgery: risk factors and prognosis. Perfusion. 2005;20(6):317–22.
105. Landoni G, Bove T, Crivellari M, Poli D, Fochi O, Marchetti C, et al. Acute renal failure after
isolated CABG surgery: six years of experience. Minerva Anestesiol. 2007;73(11):559–65.
106. Mangano CM, Diamondstone LS, Ramsay JG, Aggarwal A, Herskowitz A, Mangano
DT. Renal dysfunction after myocardial revascularization: risk factors, adverse outcomes,
and hospital resource utilization. The Multicenter Study of Perioperative Ischemia Research
Group. Ann Intern Med. 1998;128(3):194–203.
107. Wen J, Cheng Q, Zhao J, Ma Q, Song T, Liu S, et al. Hospital-acquired acute kidney injury in
Chinese very elderly persons. J Nephrol. 2013;26(3):572–9.
108. Oliveira JF, Silva CA, Barbieri CD, Oliveira GM, Zanetta DM, Burdmann EA. Prevalence
and risk factors for aminoglycoside nephrotoxicity in intensive care units. Antimicrob Agents
Chemother. 2009;53(7):2887–91.
109. Mendes CA, Cordeiro JA, Burdmann EA. Prevalence and risk factors for acute kidney injury
associated with parenteral polymyxin B use. Ann Pharmacother. 2009;43(12):1948–55.
110. Wang X, Bonventre JV, Parrish AR. The aging kidney: increased susceptibility to nephrotox-
icity. Int J Mol Sci. 2014;15(9):15358–76.
111. Li G, Lian X, Hao W, Hu W. Clinical characteristics and risk factors for mortality in older
patients with dialysis-receiving community-acquired acute kidney injury. Int J Gen Med.
2021;14:5693–701.
112. Bagshaw SM. The long-term outcome after acute renal failure. Curr Opin Crit Care.
2006;12(6):561–6.
113. Morgera S, Schneider M, Neumayer HH. Long-term outcomes after acute kidney injury. Crit
Care Med. 2008;36(4 Suppl):S193–7.
114. Coca SG. Acute kidney injury in elderly persons. Am J Kidney Dis. 2010;56(1):122–31.
115. Wu VC, Wu PC, Wu CH, Huang TM, Chang CH, Tsai PR, et al. The impact of acute kidney
injury on the long-term risk of stroke. J Am Heart Assoc. 2014;3(4)
116. Eftekhari P. Evaluation of acute kidney injury in the hospital setting. Prim Care.
2014;41(4):779–802.
117. Philips BJ, Lane K, Dixon J, Macphee I. The effects of acute renal failure on drug metabo-
lism. Expert Opin Drug Metab Toxicol. 2014;10(1):11–23.
118. Cox ZL, McCoy AB, Matheny ME, Bhave G, Peterson NB, Siew ED, et al. Adverse drug
events during AKI and its recovery. Clin J Am Soc Nephrol. 2013;8(7):1070–8.
119. American Geriatrics Society updated Beers Criteria for potentially inappropriate medication
use in older adults. J Am Geriatr Soc. 2012;60(4):616–31.
120. Gallagher P, O'Mahony D. STOPP (Screening Tool of Older Persons’ potentially inappro-
priate Prescriptions): application to acutely ill elderly patients and comparison with Beers’
criteria. Age Ageing. 2008;37(6):673–9.
121. Nawaz S, Cleveland T, Gaines PA, Chan P. Clinical risk associated with contrast angiography
in metformin treated patients: a clinical review. Clin Radiol. 1998;53(5):342–4.
426 A. Corsonello et al.
122. Subramaniam RM, Wilson RF, Turban S, et al. AHRQ comparative effectiveness reviews. In:
Contrast-induced nephropathy: comparative effectiveness of preventive measures. Rockville,
MD: Agency for Healthcare Research and Quality (US); 2016.
123. Jang JS, Jin HY, Seo JS, Yang TH, Kim DK, Kim TH, et al. Sodium bicarbonate therapy
for the prevention of contrast-induced acute kidney injury—a systematic review and meta-
analysis. Circ J. 2012;76(9):2255–65.
124. Markota D, Markota I, Starcevic B, Tomic M, Prskalo Z, Brizic I. Prevention of contrast-
induced nephropathy with Na/K citrate. Eur Heart J. 2013;34(30):2362–7.
125. Brar SS, Aharonian V, Mansukhani P, Moore N, Shen AY, Jorgensen M, et al. Haemodynamic-
guided fluid administration for the prevention of contrast-induced acute kidney injury: the
POSEIDON randomised controlled trial. Lancet. 2014;383(9931):1814–23.
126. Stevens MA, McCullough PA, Tobin KJ, Speck JP, Westveer DC, Guido-Allen DA, et al.
A prospective randomized trial of prevention measures in patients at high risk for contrast
nephropathy: results of the P.R.I.N.C.E. Study. Prevention of radiocontrast induced nephrop-
athy clinical evaluation. J Am Coll Cardiol. 1999;33(2):403–11.
127. Briguori C, Visconti G, Focaccio A, Airoldi F, Valgimigli M, Sangiorgi GM, et al. Renal
Insufficiency After Contrast Media Administration Trial II (REMEDIAL II): RenalGuard
System in high-risk patients for contrast-induced acute kidney injury. Circulation.
2011;124(11):1260–9.
128. Investigators ACT. Acetylcysteine for prevention of renal outcomes in patients under-
going coronary and peripheral vascular angiography: main results from the random-
ized Acetylcysteine for Contrast-induced nephropathy Trial (ACT). Circulation.
2011;124(11):1250–9.
129. Weisbord SD, Gallagher M, Jneid H, Garcia S, Cass A, Thwin SS, et al. Outcomes after angi-
ography with sodium bicarbonate and acetylcysteine. N Engl J Med. 2018;378(7):603–14.
130. Leoncini M, Toso A, Maioli M, Tropeano F, Villani S, Bellandi F. Early high-dose rosu-
vastatin for contrast-induced nephropathy prevention in acute coronary syndrome: Results
from the PRATO-ACS Study (Protective Effect of Rosuvastatin and Antiplatelet Therapy
On contrast-induced acute kidney injury and myocardial damage in patients with Acute
Coronary Syndrome). J Am Coll Cardiol. 2014;63(1):71–9.
131. Zhou X, Dai J, Xu X, Wang Z, Xu H, Chen J, et al. Comparative efficacy of statins for pre-
vention of contrast-induced acute kidney injury in patients with chronic kidney disease: a
network meta-analysis. Angiology. 2019;70(4):305–16.
Palliation at End of Life
29
Joanne Droney, Phoebe Wright, and Dola Awoyemi
• It can be provided in any healthcare setting and also in extended care facilities
and patients’ own homes [1, 2] with integration between services and settings
leading to improved quality of life [5].
• Palliative care can be beneficial for patients with a wide range of serious ill-
nesses, including cancer, chronic organ failure [6], dementia and progressive
neurological disorders. Palliative care has traditionally focused on patients who
There have been several recent initiatives specifically designed to enhance pallia-
tive care provision for older patients. These include the following:
• Palliative care as a core component of care for older patients with cancer [10]
• Integrated community-based palliative care for older patients with chronic non-
cancer diagnoses [11]
• Palliative care in long-term care facilities and nursing homes [12–16]
In many countries, there is an aging population, and older people are living lon-
ger with a high burden of serious health-related suffering [1, 17, 18]. Palliative care
is particularly relevant for patients with multi-morbidities, chronic illnesses with
fluctuant and unpredictable disease trajectories [19] and patients with complex clin-
ical conditions manifested in severe physical and psychosocial needs [20].
Over the next two to three decades, with major demographic changes expected in
most countries, there is likely to be a significant increase in the need for palliative
and end-of-life care for older patients, especially for those being cared for and dying
at home and in care homes [17, 21].
The paradigm for treating pain—by mouth, by the clock (on a regular basis), indi-
vidualised and with attention to detail [28]—holds true for most medications used
for symptom control. Patients suffering from pain, nausea, agitation, constipation or
respiratory secretions should have medications administered regularly to prevent
rather than to merely alleviate symptoms, with “when required” doses available.
This is particularly relevant in the frail elderly population: it has been demonstrated
that medications prescribed “when required” in residential homes are not given as
often as clinicians anticipate [29].
The preferred route for medication administration in palliative care is by mouth
[30]. However, if absorption is compromised, for example, by vomiting or intestinal
obstruction, or when swallowing is difficult in the last days of life, an alternative
route may be more appropriate, e.g. the subcutaneous route. The daily dose of medi-
cation can be given as a continuous subcutaneous infusion over 24 hours, and “when
required” medication as subcutaneous injections. A number of the medications used
frequently at the end of life can be combined within one infusion to ensure symp-
toms are managed effectively. For most medications, this route of administration is
off licence. Syringe driver compatibility charts are available to guide drug
choice [26].
This section presents a concise guide to the use of some key medications in the
management of specific symptoms in the last days of life, including pain, dyspnoea,
nausea, anxiety and agitation.
430 J. Droney et al.
Pain is common in the elderly, with around half of care home residents experiencing
pain which often remains under detected and undertreated especially in those with
dementia [29]. There is an association between pain and behavioural and psycho-
logical symptoms of dementia including agitation and anxiety [31].
For patients who are in moderate-severe pain and who have not been prescribed a
regular opioid previously, a low-dose strong opioid such as morphine should be
initiated and up-titrated, based on response, until a balance is achieved between
acceptable pain control and medication side effects. It is generally accepted that
there is little or no pharmacological indication for weak opioids in cancer pain, as
low-dose morphine (or other strong opioids) often provides better pain relief with a
lower adverse effect burden [26].
Opioids should generally be administered at regular intervals, with “as required”
doses also available for breakthrough pain. If appropriate, the oral route is prefer-
able for initial up-titration of opioids, for example, 2.5–5 mg immediate release
morphine 4 hourly by mouth. For frail older patients, it is recommended that the
starting dose is reduced by 30–50% [32] and that a more cautious approach is taken
in up-titration. For morphine, an appropriate “as required” dose is usually 1/6th of
the total 24-h morphine dose. Once the opioid has been up-titrated to a point where
good pain relief is achieved, this may be converted to either a modified release
formulation (which is administered less frequently) or, if a non-oral route is pre-
ferred, to a transdermal patch, such as a buprenorphine or fentanyl patch [33]. If
the patient is imminently dying, a subcutaneous syringe driver may be more
appropriate.
Patients who are no longer able to take oral medications can be given morphine (and
certain other strong opioids) administered as a continuous 24-h subcutaneous
syringe driver infusion. The bioavailability of subcutaneous opioids is often much
higher than oral opioids, and therefore the total 24-h subcutaneous dose will differ
from the oral equivalent. For example, 60 mg of oral morphine in a 24-h period is
approximately equivalent to 30 mg of subcutaneous morphine administered by con-
tinuous infusion over 24 h [26]. Opioids can often be combined with other
432 J. Droney et al.
The majority of patients taking morphine for cancer pain achieve good analgesia
with minimal adverse effects. Common and predictable adverse effects include con-
stipation and dry mouth, the former being largely avoidable with co-prescription of
laxatives. Other adverse effects such as nausea and vomiting are usually temporary,
following initiation of the opioid, and can be managed with a suitable antiemetic.
This may need to be added to the syringe driver if the patient is no longer able to
take oral medications. Central adverse effects, such as severe drowsiness, confu-
sion, myoclonus or hallucinations, are more concerning and suggestive of opioid
toxicity. Prompt assessment of the patient is necessary to determine the dose reduc-
tion that is required and assessment of potential precipitants, e.g. renal impairment,
infection or interacting medicines. Respiratory depression is rare if strong opioids
are titrated carefully according to individual patient response. If this occurs, the
opioid should be stopped, and the use of naloxone considered; however, this must
be judged carefully in the palliative patient population due to the risk of opioid
withdrawal syndrome and severe pain.
29 Palliation at End of Life 433
Renal and hepatic impairment is more prevalent in the older patient population
and can increase the risk of opioid toxicity. This risk varies between opioids; thus,
caution is required, and an alternate formulation or dose adjustment will often be
required [26]. Some alternate strong opioids may be indicated in specific circum-
stances, for example, fentanyl and alfentanil are safer than morphine in patients
with renal failure.
Non-opioids and adjuvants can be used to better target pain with an opioid spar-
ing intent; this is discussed in more detail elsewhere in this book.
Table 29.1 Commonly used antiemetics at end of life: mechanisms of action, indications and
cautions
Starting
“as
required”
Principle dose for
mechanism of Cautions and frail
Antiemetic action Main indication contraindications elderly
Haloperidol [42] D2 antagonist. Chemical causes of Parkinson’s disease; 0.5 mg
Acts in the nausea, e.g. renal dementia; epilepsy;
chemoreceptor failure, renal and liver
trigger zone hypercalcaemia, impairment; cardiac
medication-induced disease
Cyclizine [43] H1 and muscarinic Raised intracranial Severe heart failure; 50 mg
receptor pressure or liver disease
antagonist vestibular
symptoms
Metoclopramide D2 antagonist. Gastritis, gastric Heart disease; 10 mg
[44, 45] Acts as stasis, functional bowel obstruction;
prokinetic, also bowel obstruction Parkinson’s disease.
5HT3 receptor Avoid long-term
antagonist and treatment in older
5HT4 agonist people
Can cause acute
dystonia in older
patients
Levomepromazine D2, H1, 5HT2 and Second-line or for Parkinson’s disease; 3.125 mg
[46] muscarinic nausea of unclear dementia. Can be
antagonist aetiology sedating
D2 Dopamine type 2, H1 Histamine type 1, 5HT 5′hydroxytryptamine (serotonin)
434 J. Droney et al.
The listed antiemetics can be given orally, subcutaneously or via syringe driver
in equivalent dose (although in some specialist centres, it is advised to dose reduce
levomepromazine by 50% when converting from oral to subcutaneous).
The choice and dose of antiemetic might be influenced by several factors in older
patients. Firstly, physiological changes in the frail elderly can lead to a potential
need for dose reduction [47]. In addition, comorbidities and concurrent use of medi-
cations that might influence antiemetic choice are more common in older people.
For example, medications with dopamine antagonist activity such as metoclo-
pramide and haloperidol should be avoided in Parkinson’s disease patients and used
with caution in frail older people due to increased risk of extra-pyramidal adverse
effects [48]. In heart failure, cyclizine should be avoided. Those with pre-existing
risk factors for ventricular arrhythmia should be prescribed with caution antiemetics
that prolong the QT interval (e.g. domperidone, ondansetron or haloperidol).
Furthermore, co-prescription of some antiemetics should be avoided, for example,
cyclizine with metoclopramide, where cyclizine can be expected to block the proki-
netic action of metoclopramide.
Respiratory secretions at end of life, sometimes called the “death rattle”, are thought
to be due to an accumulation of secretions in the airways and are common in the last
hours of life, usually when the patient has become unconscious. Prior to commenc-
ing medications, non-pharmacological interventions such as repositioning can be
useful. While it is unclear if clinically assisted hydration worsens respiratory secre-
tions [49], the need for this should be regularly reviewed. In addition, it is important
to address any concerns and to provide reassurance to family members and others
present that the noisy breathing is generally not thought to be distressing to the
patient [50].
Anticholinergic medications are commonly used to reduce secretions [50]. As
older people are at increased risk, monitoring for adverse effects such as severe
xerostomia (dry mouth) and urinary retention is necessary.
Commonly used non-sedating medications include glycopyrronium and hyoscine
butylbromide. Neither is known to be more efficacious than the other [50]. Both have
poor bioavailability and therefore are given subcutaneously, either “as required” or in
a syringe driver over 24 h. The usual “as required” dose for glycopyrronium is
200–400 micrograms, with a maximum recommended dose of 1.2 mg over 24 h; for
hyoscine butylbromide, this is 20 mg with a maximum recommended dose of
60–120 mg over 24 h [51] (some specialist centres use higher maximum doses).
Hyoscine hydrobromide crosses the blood brain barrier and can cause central
adverse effects including sedation and delirium, especially in patients with renal
and liver failure. This drug can be administered subcutaneously. It is also available
as a transdermal patch, which is changed every 3 days. This may be useful in set-
tings such as nursing homes, where access to syringe drivers may be more of a chal-
lenge [52]. For patients with fluid overload secondary to heart failure, this can
29 Palliation at End of Life 435
Breathlessness is a common symptom in the last week of life. As with other symp-
toms, careful assessment is required to understand the most likely aetiology and to
ensure that any reversible causes are treated, including using supplemental oxygen
in hypoxia. Simple, instant non-pharmacological interventions can be trialled, such
as handheld or electric fans. If these are not effective, then pharmacological mea-
sures include low-dose morphine should be applied. For patients who are not immi-
nently dying, the published evidence demonstrates improvement with regular
low-dose morphine, e.g. modified release morphine, starting at 5 mg twice a day,
which can be up-titrated weekly to a maximum of 15 mg twice a day. Within these
doses, there is no evidence of excess hospital admissions or mortality [54]. The oral
route is preferred, but if a patient is thought to be in the final days of life, low-dose
morphine can be given subcutaneously for breathlessness, e.g. 5–10 mg over 24 h
via syringe driver.
Benzodiazepines are used to relieve the sensation of breathlessness especially
where there is associated anxiety [55], most commonly sublingual lorazepam or
subcutaneous midazolam. However there is no evidence in the literature to support
this approach [56].
Sometimes patients can become agitated and restless when they are imminently
dying. This is called “terminal agitation” and may be a feature of delirium at end of
life. It can also be due to uncontrolled physical symptoms or psychological or spiri-
tual distress. Reversible causes of agitation should be investigated and treated where
appropriate and when possible, e.g. hypercalcaemia and urinary retention.
The main types of medications that are used to relieve terminal agitation are
benzodiazepines and antipsychotics. It is generally not appropriate to use opioids
for this purpose [57].
Benzodiazepines, such as midazolam, can be used if anxiety is a predominant
symptom. If refractory symptoms of delirium including confusion and hallucina-
tions are predominant, then antipsychotics such as haloperidol or levomepromazine
are preferable treatment options [58]. There has been a recent move away from the
use of neuroleptics in older patients and in palliative care patients with delirium due
to a lack of high-quality evidence [59]. However, if a patient is thought to be immi-
nently dying and is exhibiting severe distress and agitation, then drug treatment can
be considered with the aim of relieving symptoms [60]. Ideally, this should be car-
ried out with specialist palliative care support.
436 J. Droney et al.
Medications may be used to relieve symptoms for patients who are approaching the
end of life. Careful assessment of cause, regular review of effect and a clear under-
standing of pharmacokinetics and pharmacodynamics of individual drugs can guide
rational and effective prescribing.
References
1. Knaul FM, Farmer PE, Krakauer EL, De Lima L, Bhadelia A, Jiang Kwete X, et al. Alleviating
the access abyss in palliative care and pain relief—an imperative of universal health coverage:
the Lancet Commission report. Lancet. 2018;391(10128):1391–454.
2. Radbruch L, De Lima L, Knaul F, Wenk R, Ali Z, Bhatnaghar S, et al. Redefining palliative
care—a new consensus-based definition. J Pain Symptom Manage. 2020;60(4):754–64.
3. Runacres F, Poon P, King S, Lustig J, Ugalde A. Examining the role of specialist palliative care
in geriatric care to inform collaborations: a survey on the knowledge, practice and attitudes of
geriatricians in providing palliative care. Age Ageing. 2021;50(5):1792–801.
4. Voumard R, Rubli Truchard E, Benaroyo L, Borasio GD, Büla C, Jox RJ. Geriatric palliative
care: a view of its concept, challenges and strategies. BMC Geriatr. 2018;18(1):1–6.
5. Bayly J, Bone AE, Ellis- C, Yaqub S, Yi D, Nkhoma KB, et al. Common elements of service
delivery models that optimise quality of life and health service use among older people with
advanced progressive conditions: a tertiary systematic review. BMJ Open. 2021;11:e048417.
6. Brännström M, Boman K. Effects of person-centred and integrated chronic heart failure
and palliative home care. PREFER: A randomized controlled study. Eur J Heart Failure.
2014;16:1142–51.
7. Van Der Steen JT, Radbruch L, Hertogh CM, De Boer ME, Hughes JC, Larkin P, et al.
White paper defining optimal palliative care in older people with dementia: a Delphi study
and recommendations from the European Association for Palliative Care. Palliat Med.
2014;28(3):197–209.
8. Temel J, Greer J, Muzikansky A, Gallagher E, Admane S, Jackson V. Early palliative care for
patients with metastatic non-small-cell lung cancer. N Engl J Med. 2010;363(8):733–42.
9. Sahlollbey N, Lee CKS, Shirin A, Joseph P. The impact of palliative care on clinical and
patient-centred outcomes in patients with advanced heart failure: a systematic review of ran-
domized controlled trials. Eur J Heart Fail. 2020;22(12):2340–6.
10. VanderWalde N, Jagsi R, Dotan E, Baumgartner J, Browner IS, Burhenn P, et al. NCCN
guidelines insights: older adult oncology, version 2.2016. J Natl Compr Cancer Network.
2016;14(11):1357–70.
11. Evans CJ, Bone AE, Yi D, Gao W, Morgan M, Taherzadeh S, et al. Community-based short-
term integrated palliative and supportive care reduces symptom distress for older people with
chronic noncancer conditions compared with usual care: a randomised controlled single-blind
mixed method trial. Int J Nurs Stud. 2021;120:103978.
438 J. Droney et al.
32. Jones MR, Ehrhardt KP, Ripoll JG, Sharma B, Padnos IW, Kaye RJ, et al. Pain in the elderly.
Vol. 20, Current pain and headache reports. Current Medicine Group LLC 1; 2016. p. 1–9.
33. Caraceni A, Hanks G, Kaasa S, Bennett MI, Brunelli C, Cherny N, et al. Use of opioid analge-
sics in the treatment of cancer pain: evidence-based recommendations from the EAPC. Lancet
Oncol. 2012;13:e58–68.
34. Bekkering GE, Soares-Weiser K, Reid K, Kessels AG, Dahan A, Treede RD, et al. Can mor-
phine still be considered to be the standard for treating chronic pain? A systematic review
including pair-wise and network meta-analyses. Curr Med Res Opin. 2011;27(7):1477–91.
35. Drewes AM, Jensen RD, Nielsen LM, Droney J, Christrup LL, Arendt-Nielsen L, et al.
Differences between opioids: pharmacological, experimental, clinical and economical per-
spectives. Br J Clin Pharmacol. 2013;75(1):60–78.
36. Riley J, Branford R, Droney J, Gretton S, Sato H, Kennett A, et al. Morphine or oxycodone
for cancer-related pain? A randomized, open-label, controlled trial. J Pain Symptom Manage.
2015;49(2):161–72.
37. Gudin J. Opioid therapies and cytochrome P450 interactions. J Pain Symptom Manage.
2012;44(6 Suppl):S4–14.
38. Gammaitoni A, Fine P, Alvarez N, McPherson LM, Bergmark S. Clinical application of opioid
equianalgesic data. Clin J Pain. 2003;19(5):286–97.
39. Fine PG, Portenoy RK. Establishing “Best Practices” for opioid rotation: conclusions of an
expert panel. J Pain Symptom Manage. 2009;38(3):418–25.
40. Stephenson J, Davies A. An assessment of aetiology-based guidelines for the manage-
ment of nausea and vomiting in patients with advanced cancer. Supportive Care Cancer.
2006;14(4):348–53.
41. Mannix K. Palliation of nausea and vomiting. In: Doyle D, Hanks G, Cherny N, Calman K,
editors. Oxford textbook of palliative medicine. 3rd ed. Oxford: Oxford University Press;
2015. p. 459–68.
42. Hardy JR, O’Shea A, White C, Gilshenan K, Welch L, Douglas C. The efficacy of haloperidol
in the management of nausea and vomiting in patients with cancer. J Pain Symptom Manage.
2010;40(1):111–6.
43. Bentley A, Boyd K. Use of clinical pictures in the management of nausea and vomiting: a
prospective audit. Palliat Med. 2001;15(3):247–53.
44. Davis MP, Hallerberg G. A systematic review of the treatment of nausea and/or vomiting in
cancer unrelated to chemotherapy or radiation. J Pain Symptom Manage. 2010;39(4):756–67.
45. Bruera E, Belzile M, Neumann C, Harsanyi Z, Babul N, Darke A. A double-blind, crossover
study of controlled-release metoclopramide and placebo for the chronic nausea and dyspepsia
of advanced cancer. J Pain Symptom Manage. 2000;19(6):427–35.
46. Dietz I, Schmitz A, Lampey I, Schulz C. Evidence for the use of Levomepromazine for symp-
tom control in the palliative care setting: a systematic review. BMC Palliat Care. 2013;12(1)
47. Gridelli C. Antiemetic therapy in older cancer patients. Aging Health. 2006;2(3):449–59.
48. Glare P, Miller J, Nikolova T, Tickoo R. Treating nausea and vomiting in palliative care: a
review. Clin Interv Aging. 2011;6(1):243–59.
49. Kingdon A, Spathis A, Brodrick R, Clarke G, Kuhn I, Barclay S. What is the impact of clini-
cally assisted hydration in the last days of life? A systematic literature review and narrative
synthesis. BMJ Support Palliat Care. 2021;11(1):68–74.
50. Wee B, Hillier R. Interventions for noisy breathing in patients near to death. Cochrane
Database Syst Rev. 2008;1
51. Wildiers H, Menten J. Death rattle: prevalence, prevention and treatment. J Pain Symptom
Manage. 2002;23(4):310–7.
52. Spiller JA, Fallon M. The use of Scopoderm in palliative care. Hospital Med. 2000;61(11):782–4.
53. Birch F, Boam E, Parsons S, Ghosh J, Johnson MJ. Subcutaneous furosemide in advanced
heart failure: service improvement project. BMJ Support Palliat Care. 2021:1–5.
54. Johnson MJ, Currow DC. Opioids for breathlessness: A narrative review. BMJ Support Palliat
Care. 2020;10(3):287–95.
440 J. Droney et al.
55. Hui D, Maddocks M, Johnson MJ, Ekström M, Simon ST, Ogliari AC, et al. Management
of breathlessness in patients with cancer: ESMO Clinical Practice Guidelines. ESMO Open.
2020;5(6):1–13.
56. Simon S, Higginson I, Booth S, Harding R, Weingärtner V, Bausewein C. Benzodiazepines
for the relief of breathlessness in advanced malignant and non-malignant diseases in adults
(Review). Cochrane Database Syst Rev. 2016;10(10)
57. Garetto F, Cancelli F, Rossi R, Maltoni M. Palliative sedation for the terminally ill patient.
CNS Drugs. 2018;32:951–61.
58. Cherny NI, Radbruch L, Chasen M, Coyle N, Charles D, Dean M, et al. European Association
for Palliative Care (EAPC) recommended framework for the use of sedation in palliative care.
Palliat Med. 2009;23(7):581–93.
59. Finucane AM, Jones L, Leurent B, Sampson EL, Stone P, Tookman A, et al. Drug therapy for
delirium in terminally ill adults. Cochrane Database Syst Rev. 2020;2020(1)
60. Hosker CMG, Bennett MI. Delirium and agitation at the end of life. BMJ (Online).
2016;353:1–6.
61. NHS Scotland. Levomepromazine [Internet]. Scottish Palliative Care Guidelines. 2021
[cited 2021 Nov 24]. https://www.palliativecareguidelines.scot.nhs.uk/guidelines/medicine-
information-sheets/levomepromazine.aspx
62. Zaporowska-Stachowiak I, Stachowiak-Szymczak K, Oduah MT, Sopata M. Haloperidol in
palliative care: Indications and risks. Biomed Pharmacother. 2020;132(October):110772.
63. “Royal College of Physicians.” National care of the dying audit for hospitals, England
Executive summary; 2014.
64. Kirk TW, Mahon MM. National Hospice and Palliative Care Organization (NHPCO) position
statement and commentary on the use of palliative sedation in imminently dying terminally ill
patients. J Pain Symptom Manage. 2010;39(5):914–23.
65. Maltoni M, Scarpi E, Rosati M, Derni S, Fabbri L, Amadori FM, et al. Palliative sedation in
end-of-life care and survival: a systematic review. J Clin Oncol. 2012;30(27)
66. Beller EM, van Driel ML, Mcgregor L, Truong S, Mitchell G. Palliative pharmacological seda-
tion for terminally ill adults. Cochrane Database Syst Rev. 2015;2017(6)
67. Cherny N. ESMO clinical practice guidelines for the management of refractory symptoms at
the end of life and the use of palliative sedation. Ann Oncol. 2014;25(July):iii143–52.
68. Bush SH, Lawlor PG, Ryan K, Centeno C, Lucchesi M, Kanji S, et al. Delirium in adult cancer
patients: ESMO Clinical Practice Guidelines. Ann Oncol. 2018 Oct;1(29):iv143–65.
69. Hosie A, Davidson PM, Agar M, Sanderson CR, Phillips J. Delirium prevalence, incidence,
and implications for screening in specialist palliative care inpatient settings: a systematic
review. Palliat Med. 2013;27(6):486–98.
70. Sampson EL, West E, Fischer T. Pain and delirium: mechanisms, assessment, and manage-
ment. Eur Geriatr Med. 2020;11(1):45–52.
Index
I N
Iatrogenic injury, 76, 78, 408, 412 Neuropsychiatric symptoms, 272, 273, 277,
Inappropriate medications, 55–56, 78, 101, 279, 282, 352
119, 120, 150, 165, 418 New York Heart Association (NYHA) class,
Inappropriate prescribing (IP), 3, 25, 264, 334
28, 47–49, 52, 54, 60, 64, 66, 76, Non pharmacologic therapy, 279
77, 99, 105, 119, 120, 149, 210, 251, Non-pharmacological treatment, 26, 376
418, 419 Nutritional deficiency, 381, 382, 387
Insomnia, 63, 64, 150, 169, 178, 281, Nutritional therapy, 386, 387
371, 375
O
L Older
Laxatives, 163, 200–209, 211, 220, 432 adults, 4–6, 9, 16–19, 21, 23, 26, 39,
59–63, 65, 66, 69–72, 75–76,
78, 81, 83, 86–88, 113, 132,
M 133, 135, 140, 143, 145–153, 162,
Major neurocognitive disorder, 269 163, 166, 167, 174, 176, 177,
Malnutrition, 133, 149, 174, 179, 218, 232, 185–197, 199, 201, 202, 209–211,
251, 252, 271, 314, 316, 317, 321, 217, 219–221, 223–225, 243, 249,
369, 381–387 252, 255, 257, 291–293, 300, 311,
Mechanical thrombectomy, 354, 355 318, 331, 333, 334, 349, 353, 369,
Medication 370, 373, 374, 376, 382, 383, 386,
discrepancy, 95, 96 388, 391, 393–395, 397–399,
error, 91, 92, 94, 97 405, 410
optimisation, 91–93, 95, 97 patients, 3, 6, 7, 10, 17–24, 26–28, 37–39,
reconciliation (MedRec), 24–26, 52, 78, 47, 49, 50, 55, 71, 72, 75–78, 81,
85, 91–98, 107, 110, 111, 114, 273 82, 91, 95, 100, 107–109, 112, 115,
review, 26–28, 48, 51–56, 60, 65, 91, 94, 120, 122, 126, 127, 144, 145, 148,
95, 98–102, 105–106, 108, 110, 149, 151–153, 174, 177, 189, 190,
111, 114, 166–169, 260, 397 195, 197, 201, 207, 217–225,
review tool, 27–28, 100 230–235, 239, 244–257, 260,
safety, 54, 91, 92, 94, 96–98 289–304, 317, 321, 322, 326, 327,
Medicines review, 98, 99, 110, 112 331–340, 351, 352, 354, 355,
Melatonin receptor agonists orexin receptor 357–359, 361, 362, 369, 371, 375,
antagonists, 392, 398 376, 387, 395, 406, 408–418,
Mobility, 20, 21, 138, 161–167, 169, 177, 188, 427–430, 433–436
289, 352, 361, 383, 384, 418 Older (cont.)
Models of pharmaceutical care people, 3, 16, 17, 24, 25, 28, 38–40, 47–56,
Mood stabilizer, 373–374 59, 61, 62, 69–78, 89, 91, 92, 95,
Morphine, 17, 18, 165, 173, 174, 219–221, 101, 109–111, 114, 119–126, 139,
223, 224, 320, 430–433, 435 161, 163, 165, 173, 177, 179, 190,
Multidisciplinary, 26, 85, 110, 112, 122, 134, 191, 196, 197, 200, 201, 211,
152, 177, 239, 244, 252, 256–260, 327, 217–219, 222, 224, 229–231, 239,
361, 362, 387 249–252, 255, 259, 260, 269, 352,
Multimorbidity, 16, 24, 28, 33, 48, 55, 353, 362, 381–384, 386, 387, 405,
56, 59, 61, 62, 72, 78, 81, 83, 86, 408, 412, 419, 429, 431, 433, 434
100, 101, 111, 131–137, 140, 141, persons, 82, 86, 87, 89, 124, 144–146, 151,
149, 165, 174, 179, 231, 239, 251, 161, 163, 165, 167, 185, 189,
312, 382, 407, 416, 417, 419 192–194, 196, 217, 218, 220,
Muscle strength, 138, 145, 162, 167, 317 245–250, 252, 253, 260, 290–304,
Muscle strength medication discrepancy 333, 369, 375, 382, 384, 406
444 Index
V Z
Vitamin D, 291, 292, 300, 302, 304, 317, 382, Z-drugs, 165, 392, 395–397
413, 414 Zoledronic acid, 298–300, 302, 303
W
Wasting, 251