Clinical Case Studies in Long-Term Care Psychiatry
Clinical Case Studies in Long-Term Care Psychiatry
Clinical Case Studies in Long-Term Care Psychiatry
Long-Term
Care Psychiatry
Navigating Common Mental Health
Challenges in Geriatric Care
Clinical Case Studies in
Long-Term Care Psychiatry
Clinical Case Studies
in Long-Term Care
Psychiatry
Navigating Common Mental Health
Challenges in Geriatric Care
Matthew Gibfried
Saint Louis University School of Medicine, Missouri
George T. Grossberg
Saint Louis University School of Medicine, Missouri
Shaftesbury Road, Cambridge CB2 8EA, United Kingdom
One Liberty Plaza, 20th Floor, New York, NY 10006, USA
477 Williamstown Road, Port Melbourne, VIC 3207, Australia
314–321, 3rd Floor, Plot 3, Splendor Forum, Jasola District Centre, New Delhi – 110025, India
103 Penang Road, #05–06/07, Visioncrest Commercial, Singapore 238467
www.cambridge.org
Information on this title: www.cambridge.org/9781108722322
DOI: 10.1017/9781108630344
© Cambridge University Press & Assessment 2025
This publication is in copyright. Subject to statutory exception and to the provisions
of relevant collective licensing agreements, no reproduction of any part may take
place without the written permission of Cambridge University Press & Assessment.
When citing this work, please include a reference to the
DOI 10.1017/9781108630344
First published 2025
A catalogue record for this publication is available from the British Library
Library of Congress Cataloging-in-Publication Data
Names: Gibfried, Matthew, author. | Grossberg, George T., author.
Title: Case studies in long-term care psychiatry : navigating common mental health challenges in geriatric care / Matthew
Gibfried, George T. Grossberg.
Description: Cambridge ; New York, NY : Cambridge University Press, 2025. | Includes bibliographical references and index. |
Summary: “A compilation of compelling real-life cases on the psychiatric care of older adults in the long-term care setting.
Cases cover the various common psychiatric disorders alongside more complex cases on psychiatric multimorbidity and
psychotropic polypharmacy. Essential reading for healthcare practitioners who work regularly with older adults”– Provided
by publisher.
Identifiers: LCCN 2024020601 (print) | LCCN 2024020602 (ebook) | ISBN 9781108722322 (paperback) | ISBN 9781108630344
(epub)
Subjects: MESH: Mental Disorders | Aged | Long-Term Care–methods | Geriatric Psychiatry–methods | Case Reports
Classification: LCC RC451.4.A5 (print) | LCC RC451.4.A5 (ebook) | NLM WT 150 | DDC 618.97/689–dc23/eng/20240627
LC record available at https://lccn.loc.gov/2024020601
LC ebook record available at https://lccn.loc.gov/2024020602
ISBN 978-1-108-72232-2 Paperback
Cambridge University Press & Assessment has no responsibility for the persistence
or accuracy of URLs for external or third-party internet websites referred to in this
publication and does not guarantee that any content on such websites is, or will remain,
accurate or appropriate.
.............................................................................................................
Every effort has been made in preparing this book to provide accurate and up-to-date information that is in accord with
accepted standards and practice at the time of publication. Although case histories are drawn from actual cases, every effort has
been made to disguise the identities of the individuals involved. Nevertheless, the authors, editors, and publishers can make no
warranties that the information contained herein is totally free from error, not least because clinical standards are constantly
changing through research and regulation. The authors, editors, and publishers therefore disclaim all liability for direct or
consequential damages resulting from the use of material contained in this book. Readers are strongly advised to pay careful
attention to information provided by the manufacturer of any drugs or equipment that they plan to use.
Contents
Preface vii
vii
viii Preface
The cases in this book are based on real-life situations. The stories, of course, are
edited for content and details that could jeopardize the anonymity of the subjects.
I purposely tried to include a wide variety of cases to touch on topics in all areas of
geriatric psychiatric pathology, including mood disorders, anxiety disorders, psychotic
disorders, major neurocognitive disorders, substance use disorders, and personality
disorders. Hopefully, the reader will find value and will be able to apply the principles
taught in these cases to their own clinical practice in the long-term care setting.
This book is written for the benefit of any provider who cares for those in long-term
care, but is also likely to provide insights into geriatric psychiatric disorders for those
who practice in the office setting as well. After finishing this book, I realized that the
experience and learning involved in writing it provided more benefit to me than anyone.
Revisiting and analyzing the cases and researching the topics were a capstone to my
fellowship experience and humbled me with knowing how much more there is still to
learn about these topics.
As mentioned earlier, I came to the field of geriatric psychiatry through a different
path than most. My training in internal medicine provided me with a wonderful
foundation for the understanding of pathophysiology and treatment of multiple organ
systems. My pull toward geriatric medicine developed over time and, eventually, I spent
the majority of my time caring for older adults in long-term care settings. It was in this
practice setting that my lack of a better understanding of major neurocognitive disorders
and other neuropsychiatric disorders felt more acute. A friend, geriatric psychiatrist, and
mentor of mine, Dr. Buntee Co, inspired me to further my career by pursuing a
fellowship in geriatric psychiatry and encouraged me to reach out to one of the pillars
of the field, Dr. George T. Grossberg, at St. Louis University School of Medicine. I am
very thankful for the support of both of these outstanding clinicians and teachers.
Dr. Grossberg pulled me into his world without hesitation and has been a consistent
source of inspiration since the day of our introduction and even before, as I tried to read
as many of his publications as possible prior to my first day of fellowship. It took me only
several hours of reading to realize that Dr. Grossberg has been more instrumental in
furthering the field of geriatric psychiatry than anyone I will likely have the pleasure to
know. His trainees are now scattered throughout the world and are training and
inspiring learners themselves just as he did. His skills as a teacher, clinician, writer,
and researcher are unmatched by anyone I have had the privilege to work with.
I would like to thank Dr. Grossberg for gently pushing me toward this opportunity to
research, write, and publish this book. He has acted as my editor, cowriter, and motivator
throughout the project. He assists me still in clinical practice as I often ask myself “What
would Dr. Grossberg do?” when confronted with difficult clinical situations.
As a side note, I would like to encourage any mid-career medical provider to consider
pursuing advanced clinical training in a subspecialty that interests them. I feel that a
mid-career fellowship has reenergized my practice and provided me with some much-
needed professional development that will benefit me throughout the rest of my career.
My family has been stoically supportive of me throughout the months of mornings
and evenings researching and writing this book. My partner, Doug, will probably be
happy to no longer have to patiently ask, “How’s the book coming?” as he has done
countless times over the past year. Our now eight- and nine-year-old kids will someday
hopefully understand why their dad was sometimes not much fun on the weekends and
seemed “super grouchy!” after spending hours typing and staring at his computer screen.
Preface ix
To those who have chosen to read this book, I hope it helps you on your journey to
become better mental health providers. Hopefully it will inspire a few to pursue even
further learning in geriatric psychiatry and pass along their knowledge to the next
generation of clinicians who will be caring for us in the future.
Matthew Gibfried, MD
Section 1 Neurocognitive Disorders
1 Sundowner Syndrome
irritable. He slept a bit better at night but would wake up in the early morning hours
asking to get out of bed into his wheelchair, at which time he would be brought close to
the nurse’s station where he would often yell at staff.
The provider started the patient on sertraline 25 mg daily, which was soon increased
to 50 mg daily. The physician recommended that the patient be brought out of his room
and encouraged to participate in activities during the daytime instead of being allowed to
stay in his room and sleep. Care staff were encouraged to bring Mr. S to the dining room
for meals instead of allowing him to take his meals in his room. This was to encourage
more socialization, combat boredom, and encourage the normalization of day/night
cycles. Melatonin 5 mg qhs was also started.
At a four-week follow-up visit, Mr. S was found to be doing somewhat better. His
mood was described as less irritable and he was more cooperative with care. His
nutritional intake improved, and his weight loss started to level off. It was noted that
care staff were taking the patient outdoors when the weather was good and he seemed to
enjoy this. His family was pleased with his improvements in neuropsychiatric symptoms
and encouraged by his increasing willingness to participate in therapies, with the goal of
Mr. S being able to participate adequately enough in his care to potentially move back
home with his family.
Teaching Points
“Sundowner syndrome” refers to a constellation of neuropsychiatric symptoms, usually
accompanied by agitation that occurs late in the day in a patient with a major neurocog-
nitive disorder (MNCD). In addition to agitation, anxiety, aggressiveness, increased
confusion, and even visual or auditory hallucinations are common. Second only to
wandering, sundowning is reported to be one of the most common disruptive behaviors
in long-term care settings [1]. Symptoms usually emerge later in the day, classically in the
late afternoon or early evening at the time of sunset. This neuropsychiatric syndrome can
be very challenging for caregivers. Sundowning is a common reason for those with
MNCDs to be moved from home to a long-term care facility. It is also one of the more
common reasons for consultation with a geriatric psychiatrist in the long-term care setting.
Sundowning and delirium share some similarities with regard to underlying causes.
Both are strongly associated with MNCDs. One key difference between sundowning and
delirium is the time of day in which symptoms occur. Sundowning typically occurs and
recurs in the late afternoon or evening hours whereas delirium can occur at any time of
the day or night.
The treatment of delirium focuses on the identification and treatment of underlying
medical issues or medications that may be contributing. In contrast, the treatment of
sundowning focuses on altering environmental factors that can contribute to
the condition.
The pathophysiology of sundowner syndrome is not fully understood, but there are
several theories about the underlying mechanisms. These theories are not mutually
exclusive.
One theory is that sundowning may be related to changes in the body’s internal clock
or circadian rhythm regulation. These rhythms are regulated by the suprachiasmatic
nucleus (SCN), which responds to light and dark stimuli to help regulate sleep–wake
cycles and other physiological processes. In individuals with dementia or other cognitive
“I’ve Fallen . . . and I Can’t Get Up” 3
impairments, the SCN may be disrupted, leading to changes in sleep patterns and other
circadian rhythms. This disruption may contribute to the development of sundowning
symptoms in the late afternoon or evening.
Another theory is that sundowning may be related to pain or fatigue that becomes
more pronounced in the late afternoon or evening. Individuals with dementia or other
cognitive impairments may have difficulty communicating their discomfort, leading to
increased agitation or confusion as a way of expressing their discomfort.
There is also evidence to suggest that sundowning may be related to changes in
neurotransmitter levels, particularly concerning the neurotransmitter acetylcholine.
Individuals with Alzheimer’s disease or other forms of MNCD have a loss of
acetylcholine-producing neurons in the brain, which contributes to changes in cognitive
function and behavior [1].
Sensory deprivation can also play a role in sundowner syndrome. Seniors in long-
term care are often lacking in stimulating daytime activities. A multidisciplinary
approach to creating individualized care plans for seniors with MNCD is important.
These care plans should allow seniors to have regular social interactions, physical
activity, family-style mealtimes, time outdoors, and religious activities.
The treatment of sundowning depends on the acuity of symptoms. All patients
should have a thorough examination of environmental factors that could be contributing
to late-day confusion and agitation and changes made to improve those environmental
factors. Medications that could be contributing to confusion should also be identified
and removed.
At the time of this writing, there has been relatively recent approval from the US Food
and Drug Administration (FDA) of the atypical antipsychotic brexpiprazole for treating
agitation due to Alzheimer’s disease[2]. There are several other pharmacologic options for
the treatment of agitation in the setting of MNCDs. For mild symptoms, several studies
have demonstrated improved outcomes for those with sundowner syndrome with mela-
tonin [1]. Melatonin levels are deficient in those with MNCDs (especially Alzheimer’s
disease). Melatonin is generally well tolerated and may be worth trying in those with mild
symptoms. In those patients with more severe symptoms, including severe agitation or
psychosis, a trial of a second- or third-generation antipsychotic may be warranted [1].
Antipsychotics are used off-label and should be used with caution and in the smallest dose
for the shortest possible duration. Regular efforts to decrease the dosage of antipsychotics
are also needed once symptoms are controlled. Cholinesterase inhibitors such as donepezil
are known to improve behavioral disturbances in patients with dementia but there are
conflicting data on the effect of cholinesterase inhibitors on sundowner syndrome [1].
Take-Home Points
– Antipsychotics are used off-label and should be used with caution and in the smallest dose for the
shortest possible duration. Regular efforts to decrease the dosage of antipsychotics are also needed
once symptoms are controlled.
1 Sundowner Syndrome
irritable. He slept a bit better at night but would wake up in the early morning hours
asking to get out of bed into his wheelchair, at which time he would be brought close to
the nurse’s station where he would often yell at staff.
The provider started the patient on sertraline 25 mg daily, which was soon increased
to 50 mg daily. The physician recommended that the patient be brought out of his room
and encouraged to participate in activities during the daytime instead of being allowed to
stay in his room and sleep. Care staff were encouraged to bring Mr. S to the dining room
for meals instead of allowing him to take his meals in his room. This was to encourage
more socialization, combat boredom, and encourage the normalization of day/night
cycles. Melatonin 5 mg qhs was also started.
At a four-week follow-up visit, Mr. S was found to be doing somewhat better. His
mood was described as less irritable and he was more cooperative with care. His
nutritional intake improved, and his weight loss started to level off. It was noted that
care staff were taking the patient outdoors when the weather was good and he seemed to
enjoy this. His family was pleased with his improvements in neuropsychiatric symptoms
and encouraged by his increasing willingness to participate in therapies, with the goal of
Mr. S being able to participate adequately enough in his care to potentially move back
home with his family.
Teaching Points
“Sundowner syndrome” refers to a constellation of neuropsychiatric symptoms, usually
accompanied by agitation that occurs late in the day in a patient with a major neurocog-
nitive disorder (MNCD). In addition to agitation, anxiety, aggressiveness, increased
confusion, and even visual or auditory hallucinations are common. Second only to
wandering, sundowning is reported to be one of the most common disruptive behaviors
in long-term care settings [1]. Symptoms usually emerge later in the day, classically in the
late afternoon or early evening at the time of sunset. This neuropsychiatric syndrome can
be very challenging for caregivers. Sundowning is a common reason for those with
MNCDs to be moved from home to a long-term care facility. It is also one of the more
common reasons for consultation with a geriatric psychiatrist in the long-term care setting.
Sundowning and delirium share some similarities with regard to underlying causes.
Both are strongly associated with MNCDs. One key difference between sundowning and
delirium is the time of day in which symptoms occur. Sundowning typically occurs and
recurs in the late afternoon or evening hours whereas delirium can occur at any time of
the day or night.
The treatment of delirium focuses on the identification and treatment of underlying
medical issues or medications that may be contributing. In contrast, the treatment of
sundowning focuses on altering environmental factors that can contribute to
the condition.
The pathophysiology of sundowner syndrome is not fully understood, but there are
several theories about the underlying mechanisms. These theories are not mutually
exclusive.
One theory is that sundowning may be related to changes in the body’s internal clock
or circadian rhythm regulation. These rhythms are regulated by the suprachiasmatic
nucleus (SCN), which responds to light and dark stimuli to help regulate sleep–wake
cycles and other physiological processes. In individuals with dementia or other cognitive
“I’ve Fallen . . . and I Can’t Get Up” 3
impairments, the SCN may be disrupted, leading to changes in sleep patterns and other
circadian rhythms. This disruption may contribute to the development of sundowning
symptoms in the late afternoon or evening.
Another theory is that sundowning may be related to pain or fatigue that becomes
more pronounced in the late afternoon or evening. Individuals with dementia or other
cognitive impairments may have difficulty communicating their discomfort, leading to
increased agitation or confusion as a way of expressing their discomfort.
There is also evidence to suggest that sundowning may be related to changes in
neurotransmitter levels, particularly concerning the neurotransmitter acetylcholine.
Individuals with Alzheimer’s disease or other forms of MNCD have a loss of
acetylcholine-producing neurons in the brain, which contributes to changes in cognitive
function and behavior [1].
Sensory deprivation can also play a role in sundowner syndrome. Seniors in long-
term care are often lacking in stimulating daytime activities. A multidisciplinary
approach to creating individualized care plans for seniors with MNCD is important.
These care plans should allow seniors to have regular social interactions, physical
activity, family-style mealtimes, time outdoors, and religious activities.
The treatment of sundowning depends on the acuity of symptoms. All patients
should have a thorough examination of environmental factors that could be contributing
to late-day confusion and agitation and changes made to improve those environmental
factors. Medications that could be contributing to confusion should also be identified
and removed.
At the time of this writing, there has been relatively recent approval from the US Food
and Drug Administration (FDA) of the atypical antipsychotic brexpiprazole for treating
agitation due to Alzheimer’s disease[2]. There are several other pharmacologic options for
the treatment of agitation in the setting of MNCDs. For mild symptoms, several studies
have demonstrated improved outcomes for those with sundowner syndrome with mela-
tonin [1]. Melatonin levels are deficient in those with MNCDs (especially Alzheimer’s
disease). Melatonin is generally well tolerated and may be worth trying in those with mild
symptoms. In those patients with more severe symptoms, including severe agitation or
psychosis, a trial of a second- or third-generation antipsychotic may be warranted [1].
Antipsychotics are used off-label and should be used with caution and in the smallest dose
for the shortest possible duration. Regular efforts to decrease the dosage of antipsychotics
are also needed once symptoms are controlled. Cholinesterase inhibitors such as donepezil
are known to improve behavioral disturbances in patients with dementia but there are
conflicting data on the effect of cholinesterase inhibitors on sundowner syndrome [1].
Take-Home Points
– Antipsychotics are used off-label and should be used with caution and in the smallest dose for the
shortest possible duration. Regular efforts to decrease the dosage of antipsychotics are also needed
once symptoms are controlled.
retaining knowledge from one therapy session to another. Mr. T scored 18 out of 30 on
his Saint Louis University mental status (SLUMS) exam. He was also found to have
significant peripheral neuropathy in his feet and legs. Over the course of the next several
weeks, it became clear that Mr. T was not going to be able to return to an independent
living situation. Mr. T’s daughter assisted social services in making arrangements for
long-term care.
Mr. T’s long-term care provider felt that Mr. T’s MNCD was related to chronic
alcohol use disorder (AUD).
Teaching Points
The majority of cases of MNCDs can be classified as related to Alzheimer’s disease,
vascular mechanisms, or Lewy Body dementia. Major neurocognitive disorders related to
chronic alcohol use may be underrecognized as there are no definitive diagnostic criteria.
The prevalence of the condition varies largely depending on the demographics of the
population being studied, as alcohol consumption patterns vary based on social and
ethnic patterns of alcohol use. It is estimated that 10–24% of those who have chronic
AUD will develop dementia [1]. Studies have shown that alcohol-related dementia
(ARD) is responsible for about 10% of early-onset dementia and about 1% of late-
onset MNCDs [2].
The pathophysiology of ARD is complex and at present not well understood.
Suggested factors likely to be involved include direct neurotoxicity of alcohol, chronic
oxidative stress, neuroinflammation, vitamin deficiencies, alcohol-related impacts on
neurotransmitter pathways, and vascular effects [3]. See Box 2.1.
There are psychosocial comorbidities to AUD that likely influence the development
of dementia at a younger age than in non-alcohol-abusing cohorts. Those with AUD
often experience social isolation, financial difficulties, relationship problems, and
Box 2.1 Suggested mechanisms influencing the development of MNCD in chronic AUD
1. Direct neurotoxicity of alcohol. Alcohol can disrupt cell membrane integrity and alter
ionic balance within neurons. This can lead to cell damage or death [3].
2. Oxidative stress related to alcohol. The metabolism of alcohol creates reactive oxygen
species as byproducts such as superoxide radicals and hydrogen peroxide. Heavy
production of these substances can overwhelm antioxidant defense mechanisms and
cause oxidative damage to cell membranes, intracellular proteins, and DNA.
3. Alcohol-related neuroinflammation. Alcohol can activate immune cells in the central
nervous system (CNS) and lead to the release of pro-inflammatory cytokines, which can
cause damage to cells within the CNS [4].
4. Vitamin deficiencies. Chronic heavy alcohol use can lead to deficiencies of thiamine,
folate, and pyridoxine. These deficiencies can lead to dysfunctional cellular energy
metabolism, impaired DNA synthesis and repair, and neurotransmitter synthesis.
5. Alcohol-related effects on neurotransmitter pathways. Alcohol negatively alters some
key neurotransmitter systems.
6. Central nervous system vascular dysfunction related to chronic alcohol use. Chronically
high levels of alcohol use can cause disruptions of the blood–brain barrier, cerebral
blood flow irregularities, endothelial dysfunction, and hypertension-related small
vessel disease of the brain [5].
“I Think I Hit My Head” 7
housing insecurity that may contribute to earlier cognitive decline. Those with AUD are
less likely to adhere to medical treatments for conditions such as hypertension, diabetes,
and cerebrovascular disease, causing earlier CNS-related complications of these condi-
tions. Those with AUD are also much more likely to have alcohol-related head injuries
from falls, which is known to increase the risk of early progressive cognitive impairment.
Unlike some other MNCDs, there are no specific cerebrospinal fluid (CSF) or
imaging findings that are definitive for alcohol-related MNCD. Diagnosis is usually
clinical. Those with AUD-related MNCD are more likely to have an earlier onset of
dementia and the cognitive impairment will be in the setting of multiple other associated
medical and psychosocial comorbidities of AUD [1]. Some other common conditions
associated with chronic AUD are shown in Box 2.2.
Although uncommon, providers may be faced with the situation of a patient in long-
term care who continues to intermittently abuse alcohol. Cravings for alcohol may
remain long after admission and those patients with relatively preserved cognition
and/or help from enabling friends or family may have access to alcohol while there.
Naltrexone provides a generally well-tolerated pharmacologic option for blunting the
pleasurable effects of alcohol in those with AUD. It acts through blockade of the mu-
opioid receptors and therefore should not be administered to those who require opiate
medications. Acamprosate is a good pharmacologic alternative for those patients who
require intermittent or regular opiate use [7].
There are no specific pharmacologic treatments for alcohol-related MNCD. Clinical
trials exploring the efficacy of cholinesterase inhibitors specifically in ARD are limited,
and the existing data on their effectiveness on this condition are inconclusive. Treatment
is largely symptomatic and focuses on the often-seen medical and psychiatric comorbid-
ities that are seen with the condition. Abstinence from alcohol is the most critical factor
8 Neurocognitive Disorders
in slowing the further decline in alcohol-related MNCD. By the time a patient ends up in
long-term care, they have often been abstaining from alcohol for some time due to a lack
of access from being in a highly controlled care setting. Cognitive rehabilitation with
occupational therapy or speech therapy can produce some improvements in functional-
ity and nutritional support is important. Those with ARD may have a long history of
very poor nutrition. Psychiatric comorbidities such as depression and anxiety are
common and should be treated similarly to those without a history of AUD. Social
support and engagement can improve the quality of life in these patients.
The prognosis of ARD can be difficult to predict as there are many confounding
variables that may influence outcomes. Those who have early recognition of the condi-
tion with early interventions are likely to fare better than those who present with more
severe cognitive impairment in a later stage of the disease. Conversely, those who present
with severe cognitive impairment at a younger age are more likely to have more
aggressive cognitive decline. Those who cease consumption of alcohol clearly do better
than those who continue to abuse alcohol and patients with fewer medical comorbidities
have a longer life expectancy. In addition, those in a more controlled and supportive
environment such as long-term care or assisted living with greater support of healthy
living and less availability of alcohol are likely to have improved outcomes.
There are certain difficult situations that may arise in the care of those with early
ARD in the assisted living or long-term care environment. Those with less severe
cognitive impairments are often still able to direct their own care and may have the
ability to obtain alcohol independently or via the help of others. Balancing the autonomy
of the resident to potentially make bad choices with a facility’s responsibility to provide a
safe and healthy environment for the resident and other residents can be a difficult
tightrope to navigate. The consequences of continued heavy alcohol use in one already
experiencing cognitive and health declines will often progress to the point where the
individual will need a surrogate decision-maker to facilitate placement in a more
controlled environment, such as long-term care or memory care. Once active drinking
has stopped, these patients may maintain relative medical and cognitive stability or
experience only slow continued declines in cognitive abilities.
Take-Home Points
– Chronic heavy alcohol use is toxic to the brain via multiple mechanisms.
– Labs and imaging are not specific for alcohol-related MNCD. Diagnosis is usually via history and
clinical findings.
– Alcohol-induced MNCD can be partially reversible with abstinence, but the degree of reversibility
depends on the severity of pathology.
– Once alcohol use has stopped there can be a long period of cognitive stability.
3. Zhang, K., & Luo, J. (2019). Role of MCP- 7. Murphy, C. E., Wang, R. C., Montoy, J.
1 and CCR2 in alcohol neurotoxicity. C., Whittaker, E., & Raven, M. (2022).
Pharmacological Research, 139, 360. Effect of extended-release naltrexone on
https://doi.org/10.1016/j.phrs.2018.11 alcohol consumption: A systematic
.030 review and meta-analysis. Addiction,
4. King, J. A., Nephew, B. C., Choudhury, 117 (2), 271–281. https://doi.org/10.1111/
A., Poirier, G. L., Lim, A., & Mandrekar, add.15572
P. (2020). Chronic alcohol-induced liver
injury correlates with memory deficits: Further Reading
Role for neuroinflammation. Alcohol Kaldy, J. (2023). Alcohol use in long-term care
(Fayetteville, N.Y.), 83, 75–81. https://doi communities: Juggling choice and safety.
.org/10.1016/j.alcohol.2019.07.005 Caring for the Ages, 24 (1), 1. https://doi
5. Sachdeva, A., Chandra, M., Choudhary, .org/10.1016/j.carage.2022.12.013
M., Dayal, P., & Anand, K. S. (2016). Lohoff, F. W. (2020). Pharmacotherapies and
Alcohol-related dementia and personalized medicine for alcohol use
neurocognitive impairment: A review disorder: A review. Pharmacogenomics,
study. International Journal of High-Risk 21 (15), 1117–1138. https://doi.org/10
Behaviors & Addiction, 5 (3), e27976 .2217/pgs-2020-0079
https://doi.org/10.5812/ijhrba.27976
Sullivan, E. V., & Pfefferbaum, A. (2019).
6. MacKillop, J., Agabio, R., Feldstein- Brain-behavior relations and effects of
Ewing, S., Heilig, M., Kelly, J. F., Leggio, aging and common comorbidities in
L., Lingford-Hughes, A., Palmer, A., alcohol use disorder: A review.
Parry, C., Ray, L., & Rehm, J. (2022). Neuropsychology, 33 (6), 760. https://doi
Hazardous drinking and alcohol use .org/10.1037/neu0000557
disorders. Nature Reviews. Disease
Primers, 8 (1), 80. https://doi.org/10.1038/
s41572–022-00406-1
Section 1 Neurocognitive Disorders
Mrs. S was a 78-year-old resident of a medium-sized skilled nursing facility. She had
been a resident of the facility for four years following a fall at home. Nursing staff at the
facility requested that the consulting psychiatrist see the patient after she had returned
from hospital where she was evaluated after a fall with a scalp laceration. Since returning
from the emergency department Mrs. S had not been acting as usual. She had been
showing personality changes. Prior to her fall, she could be described as fairly quiet but
pleasant. She enjoyed social functions in the nursing facility and was generally liked by
other residents and nursing staff alike. She had a particular affinity for music and her
favorite activities involved musical performers who would come to the facility once or
twice a week. Mrs. S loved to play the piano and would often entertain others in the
facility after evening meals with her piano playing and singing.
After her return from the emergency room following her fall three weeks prior, Mrs. S
seemed to have little interest in social activities. She had stopped attending most musical
activities and had stopped playing music for other residents even after much encourage-
ment from her friends. Her friends and staff members had noted that Mrs. S was now
described as often short-tempered and irritable with others. Mrs. S’s daughter was also
concerned about these changes. “It’s like she’s a different person, what happened to my
mom!” her daughter exclaimed during the provider’s first visit with the patient.
On review of Mrs. S’s chart before the meeting, it was noted that she had a history of
obesity class 3 (BMI 43), impaired mobility, hypertension, hyperlipidemia, and psoriasis.
She had polio as a young girl which left her with shortening of her left lower extremity,
scoliosis, and lifelong issues with impaired mobility. For most of her life, she was fully
mobile with a cane or bilateral hand crutches but as she reached her 50s and 60s she
began gaining weight. Her weight gain led to a slow steady decline in mobility. Prior to
her most recent fall, she was dependent mostly on a motorized scooter for mobility but
was still able to self-transfer from her bed to her scooter with the aid of her hand
crutches.
A review of her chart also indicated that Mrs. S had several hospital visits for falls in
the past year alone. None had resulted in serious injury, only scalp contusions or
lacerations. The nursing staff mentioned that Mrs. S may have lost consciousness for
“a while” after her most recent fall. Her falls always happened when she tried to transfer
herself to her motorized scooter or while transferring on and off the commode. She had a
CT brain scan after her latest fall that showed only mild age-related small vessel ischemic
changes. There was no comment on atrophy or subdural hematoma. A review of recent
labs indicated that the patient’s comprehensive metabolic panel (CMP), complete blood
count (CBC), thyroid studies, b12, and folate levels were all within normal limits.
10
“It’s like She’s a Different Person” 11
Mrs. S’s daughter was happy to be present during the initial psychiatric examination.
Mrs. S was not very forthcoming during the interview. She could answer simple direct
questions, but her daughter was forced to give most of the history. Mrs. S had no history
of mental health issues. She was described as only mildly forgetful at times, but this had
changed in recent weeks. Mrs. S now seemed unable to remember visits with her
daughter from one week to the next and had little interest in gossiping about her
extended family members or discussing her favorite soap operas as she used to do. She
seemed to still be eating well. She slept well at night and indeed seemed to be sleeping
more during the day as well.
Mrs. S scored a two out of a possible five on a mini-cog exam. Her mental status
exam revealed her to be awake and alert. She was oriented to person and place but not to
month or year. Her hygiene was fair to poor, her hair was disheveled, and she was
wearing her nightgown that was littered with food stains at 2 p.m. Her mood seemed
somewhat depressed and mildly irritable. Her affect was flat. She did not show any signs
of hallucinations and denied feeling suicidal. Her behavior was calm. Her neurologic
exam was consistent with her history of post-polio syndrome with weakness in her left
leg, but no other focal findings were seen.
The patient and her daughter were counseled on the topic of neuropsychiatric
complications of traumatic brain injury (TBI). They were informed that the diagnosis
was that of depression following a TBI sustained during her last fall. It was recom-
mended that the patient begin an antidepressant in hopes of improving her mood,
energy, and social withdrawal. She started on fluoxetine 10 mg daily which was increased
to 20 mg daily after two weeks. On a six-week follow-up visit, Mrs. S was doing
somewhat better. Her daughter stated that Mrs. S was more pleasant and getting involved
in more day-to-day activities at the facility. Her ability to play the piano had not quite
returned to her pre-fall baseline; she would sit down at the piano and become frustrated
after five or ten minutes when she could not seem to remember the songs that she had
been playing for many years. Despite this, she still enjoyed listening to the volunteer
musicians from the community who played at the facility. Mrs. S was finishing a course
of physical and occupational therapy that included some cognitive retraining. Her
cognition and ability to transfer seemed to be improving.
Teaching Points
Traumatic brain injury is more common in older adults than in any other age group. It is
the most common fall-related injury among adults over the age of 65 [1]. Hospitalization
rates for TBI increase exponentially with age. Rates of hospitalization for TBI are three
times higher in those over 75 when compared to patients 65–74 years of age [1].
Traumatic brain injury is not only considered an acute condition but a chronic one as
well. Most TBIs in low- to moderate-income countries are attributed to road traffic
accidents while the highest numbers of TBIs in the US and other high-income countries
are due to falls, especially in those over the age of 65. Older patients are generally
considered to have poorer outcomes with more severe cognitive and functional impair-
ments from TBI than younger patients [2].
The pathogenesis of TBI involves both primary and secondary injury mechanisms.
Primary mechanisms involve mechanical injury that occurs from the physical forces that
occur during the fall, including acceleration, deceleration, and direct impact. These can
12 Neurocognitive Disorders
cause contusions of brain tissues and surrounding structures. These mechanical forces
also cause diffuse axonal injury from widespread damage to axons caused by shearing
forces from acceleration and deceleration. These acute injuries often lead to short-term
brain dysfunction resulting in concussion. Secondary injury mechanisms are the subse-
quent processes that follow the primary injury and can exacerbate damage to the brain.
These mechanisms can evolve, sometimes hours to days after the initial trauma. Box 3.1
lists some of the secondary injury mechanisms of TBI [3].
Older adults are at higher risk of worse outcomes following traumatic brain injuries.
Older adults are known to have age-related changes in brain structure such as brain
atrophy and reduced blood flow, which make them more susceptible to injury. These
changes can result in a decreased ability to recover from brain trauma. Older adults also
often have pre-existing medical conditions which can complicate recovery from TBI.
These comorbidities include higher incidences of hypertension, diabetes, and cerebro-
vascular disease. Medications more likely to be utilized in older adults can also increase
the risk of TBI. These include medications that can increase the risk of intracerebral
hemorrhage or microhemorrhages after injury. Older adults generally have slower
healing following injury and may have a lower cognitive reserve, which lowers their
ability to cognitively compensate for the effects of brain injury. The prevalence of
coexisting neuropsychiatric disorders such as depression, anxiety, and neurodegenerative
diseases is also higher in older adults. These comorbidities can complicate recovery.
Seniors may have more limited access to post-injury specialized care. The increasing
frailty of older adults can affect their ability to tolerate the physical and emotional stress
of a TBI and its associated rehabilitation [3,4].
The treatment of traumatic brain injuries in older adults should consider the unique
needs and challenges of this population. Hospital-level care focuses on stabilization and
treating any acute issues such as bleeding or swelling of the brain. Rehabilitation is a
critical component of TBI treatment for older adults. This includes physical therapy,
“It’s like She’s a Different Person” 13
Box 3.2 Common neuropsychiatric conditions that may result from TBI
Post-concussion Syndrome:
Post-concussion syndrome (PCS) is a common diagnosis following mild to moderate
TBIs. It is characterized by a constellation of symptoms, including headaches, dizziness,
fatigue, irritability, memory problems, and difficulty concentrating.
Mood Disorders:
Depression: TBI can trigger depressive symptoms, which may include persistent
sadness, loss of interest in activities, changes in appetite, and sleep disturbances.
Anxiety: Generalized anxiety, panic disorder, and specific phobias may develop or
worsen after TBI.
Bipolar disorder: Some patients may experience mood swings and manic or hypomanic
episodes following TBI.
Psychotic Disorders:
In some cases, TBI can trigger or exacerbate psychotic symptoms, leading to a
diagnosis of schizophrenia or other psychotic disorders.
Personality Changes:
TBI can lead to alterations in personality, such as apathy, reduced impulse control, and
diminished empathy.
Sleep Disorders:
Insomnia, excessive daytime sleepiness, and sleep-related breathing disorders can
develop or worsen after TBI.
Neurodegenerative Disorders:
One study showed that TBI in older adults is associated with a 44% increased risk of
Parkinson’s disease and a 26% increase of MNCDs in general [7].
Take-Home Points
– Traumatic brain injury is more common in older adults than in any other age group. It is the most
common fall-related injury among adults over the age of 65.
– The pathogenesis of TBI involves both primary and secondary injury mechanisms. Older adults are
at higher risk of worse outcomes following traumatic brain injuries.
– Traumatic brain injuries can lead to a wide range of neuropsychiatric symptoms and diagnoses
both in the acute and long term. These can include mood disorders, cognitive impairment, and
increased risk of MNCD and Parkinson’s disease.
– The severity of TBI in older adults lies in their vulnerability, the potential for worse outcomes, and
the need for tailored care and prevention strategies to address the unique challenges of
this population.
4. Chauhan, A. V., Guralnik, J., Sorkin, J. D., C. M., & Yaffe, K. (2015). Traumatic
Badjatia, N., & Albrecht, J. S. (2022). brain injury in later life increases risk for
Repetitive traumatic brain injury among Parkinson’s disease. Annals of Neurology,
older adults. The Journal of Head Trauma 77, 987–995.
Rehabilitation, 37 (4), E242. https://doi
.org/10.1097/HTR.0000000000000719
Further Reading
5. Brett, B. L., Kramer, M. D., Whyte, J., Cash, A., & Theus, M. H. (2020). Mechanisms
McCrea, M. A., Stein, M. B., Giacino, of blood–brain barrier dysfunction in
J. T., Sherer, M., Markowitz, A. J., traumatic brain injury. International
Manley, G. T., Nelson, L. D., Journal of Molecular Sciences, 21 (9).
Investigators, T., Adeoye, O., Badjatia, N., https://doi.org/10.3390/ijms21093344
Boase, K., Barber, J., Bodien, Y., Bullock,
M. R., Chesnut, R., Corrigan, J. D., & Perry, D. C., Sturm, V. E., Peterson, M. J.,
Zafonte, R. (2021). Latent profile analysis Pieper, C. F., Bullock, T., Boeve, B. F.,
of neuropsychiatric symptoms and Miller, B. L., Guskiewicz, K. M., Berger,
cognitive function of adults 2 weeks after M. S., Kramer, J. H., & Welsh-Bohmer,
traumatic brain injury: Findings from the K. A. (2016). Association of traumatic brain
TRACK-TBI study. JAMA Network Open, injury with subsequent neurological and
4 (3), e213467. https://doi.org/10.1001/ psychiatric disease: A meta-analysis.
jamanetworkopen.2021.3467 Journal of Neurosurgery, 124 (2),
511–526. https://doi.org/10.3171/2015.2
6. Ahmed, S., Venigalla, H., Mekala, H. M., .JNS14503
Dar, S., Hassan, M., & Ayub, S. (2017).
Traumatic brain injury and Torregrossa, W., Raciti, L., Rifici, C., Rizzo, G.,
neuropsychiatric complications. Indian Raciti, G., Casella, C., Naro, A., & Calabrò,
Journal of Psychological Medicine, 39 (2), R. S. (2023). Behavioral and psychiatric
114–121. https://doi.org/10.4103/0253- symptoms in patients with severe traumatic
7176.203129 brain injury: A comprehensive overview.
Biomedicines, 11 (5), 1449. https://doi.org/
7. Gardner, R. C., Burke, J. F., 10.3390/biomedicines11051449
Nettiksimmons, J., Goldman, S., Tanner,
Section 1 Neurocognitive Disorders
Ms. S, a 79-year-old woman residing in a nursing home, was referred to psychiatry for
evaluation of suspected depression. She had a history of memory problems, disorien-
tation at times, recurrent falls, and sleep disturbances beginning two to three years ago
and had been progressively getting worse. Her most recent fall caused her to have a
bruise on her face but no fracture, and for two weeks she was increasingly withdrawn,
staying in her room with mild impairment in appetite. On evaluation, the resident’s
main complaint was fatigue; she stated, “I am tired all the time.” Her medical history was
positive for hypertension, peripheral vascular disease, osteoarthritis of the knees, and
carpal tunnel syndrome.
A review of systems revealed dysuria. She presented mentally slowed, with flat affect, and
moderately disoriented to time and place. Her SLUMS score was 12/30. Physical examin-
ation revealed a low-frequency tremor of the arms and a slow gait. Laboratory results were
within normal range and urinalysis revealed a bladder infection, which was treated with
antibiotics. Ms. S was taking amlodipine, acetaminophen, and lorazepam 0.5 mg three times
a day. To rule out chronic subdural hematoma, the physician recommended an emergency
CT scan of the head, which showed old diffuse ischemic white matter changes and mild
cortical atrophy. The lorazepam was gradually tapered and discontinued over two weeks.
Ms. S was started on donepezil 5 mg once daily. Family and staff were counseled to have her
walk regularly, as she had always enjoyed walking. After one month, she was sleeping better
and her dysuria had cleared up, but she still complained of fatigue. She had tolerated
donepezil well so far without developing any nausea or vomiting and her tremor had not
worsened. The donepezil was increased to 10 mg daily. Water painting was added to her
activity schedule, as Ms. S had shown interest in this activity when she was in an adult day
program. After another month, she appeared to be doing much better. She still complained
of fatigue, but less often. Her family felt she was more alert and interactive. Her repeat
SLUMS score was 15. Final diagnosis of probable dementia with Lewy bodies (DLB)
was given.
Teaching Points
It is important to note that adverse effects of benzodiazepines, such as fatigue, may take
several weeks to improve after the drug is discontinued. Also, a UTI may manifest as
fatigue, depression, and/or increased confusion. Often, there are multiple causes of
symptoms and disability, with untreated MNCD being one of them. In this case,
treatment of the UTI, discontinuation of lorazepam, the institution of cholinesterase
inhibitor (ChEi) therapy, and aggressive use of strength-based personalized psychosocial
16
“I Am Tired All the Time” 17
sensory spiritual environmental initiatives with creative engagement helped improve the
resident’s quality of life.
Dementia with Lewy bodies is a neurodegenerative disorder characterized by the
presence of Lewy bodies, which are abnormal protein deposits, in certain areas of the
brain. These Lewy bodies are composed mainly of alpha-synuclein protein and their
accumulation in the brain leads to the death of brain cells, causing a decline in cognitive
and motor functions. Dementia with Lewy bodies is the second most common form of
dementia after Alzheimer’s disease, accounting for about 15% of all dementia cases. The
accumulation of Lewy bodies (α-synuclein neuronal inclusions) and cell death is also the
key pathophysiologic finding in Parkinson’s disease. Up to 80% of those diagnosed with
Parkinson’s disease go on to develop DLB.
The symptoms of DLB can be variable and fluctuate from day to day, making it
challenging to diagnose. The most common symptoms include cognitive impairment,
such as difficulties with attention, memory, and problem-solving, as well as visual
hallucinations, fluctuations in alertness and attention, and motor symptoms, such as
muscle rigidity, tremors, and difficulty with balance and gait. Some individuals with DLB
may also experience sleep disturbances, depression, and anxiety.
Dementia with Lewy bodies can be difficult to diagnose because there is a great deal
of overlap with other neurodegenerative disorders such as vascular dementia,
Parkinson’s disease, and Alzheimer’s disease. As with all potentially new MNCD diag-
noses, a thorough medical history, physical exam, and neurologic testing are important
in making a diagnosis. Imaging modalities such as MRI and dopamine active transporter
(DAT) positron emission tomography (PET) scans can be helpful in making an accurate
diagnosis and ruling out other possible causes of cognitive impairment.
There are central features, core features, suggestive features, and supportive features
that can be used to aid in the diagnosis of DLB. Central features are required for possible
or probable diagnosis of DLB. These include progressive dementia severe enough to
interfere with normal social or occupational function and deficits on tests of attention,
executive function, and visuospatial ability. Core features include fluctuating cognition,
recurrent visual hallucinations, and spontaneous Parkinsonism. Two core features are
required for probable DLB and one for possible diagnosis. Suggestive features of DLB
include rapid eye movement (REM) sleep behavior disorder, severe sensitivity to anti-
psychotics, and low dopamine transporter uptake in the basal ganglia. Any suggestive
feature with at least one core feature defines probable DLB. Any suggestive feature in the
absence of core features defines possible DLB. Supportive features can be commonly seen
in DLB but do not have specificity for the disease. These features can include severe
autonomic dysfunction, systematized delusions, depression, and unexplained loss of
consciousness, among others.
REM sleep behavior disorder (RBD) is common in DLB and Parkinson-related
dementia. It is a sleep disorder in which a person physically acts out their dreams during
the REM stage of sleep. Normally, during REM sleep, the body’s muscles are temporarily
paralyzed, preventing movement during dreams. However, in individuals with RBD, this
muscle paralysis is incomplete or absent, leading to physical movements and vocaliza-
tions during sleep. The movements associated with RBD can range from the simple, such
as kicking or punching, to the complex and potentially dangerous, such as jumping out
of bed or running around the room. This can pose a risk of injury to the person with
RBD as well as their bed partner.
18 Neurocognitive Disorders
Take-Home Points
Dementia with Lewy bodies is a neurodegenerative disorder characterized by the presence of Lewy
bodies, which are abnormal protein deposits, in certain areas of the brain. It is the second most
common form of MNCD after Alzheimer’s disease, accounting for about 15% of all dementia cases.
Unfortunately, there are no curative treatments for DLB, and treatment is mostly symptomatic.
Medications such as cholinesterase inhibitors can provide benefits and help improve cognition and
reduce hallucinations in some individuals with DLB.
There are central features, core features, suggestive features, and supportive features that can be
used to aid in the diagnosis of DLB.
Second- or third-generation antipsychotics are preferred over first-generation antipsychotics in
those with DLB. Quetiapine is probably the best choice if an antipsychotic is needed for neurop-
sychiatric symptoms in patients with DLB.
“I Am Okay”
Case
5 Frontotemporal Dementia
Teaching Points
Frontotemporal dementia is the name of a group of dementias that primarily involve the
frontal and temporal lobes of the brain. They are progressive neurodegenerative dis-
orders that are typically diagnosed in individuals under the age of 65. Major declines in
cognition, behavior, and language are seen. Frontotemporal dementias and their variants
are the second most common type of dementia diagnosed in those under 65 years of age
and the third most common type of MNCD overall, behind Alzheimer’s disease and
dementia with Lewy bodies. Different epidemiologic studies have shown relatively wide
differences in estimations of the prevalence of the disease. Estimating the prevalence of
20
“I Am Okay” 21
FTD can be difficult as there is a good deal of overlap in symptomatology between FTD
and other forms of major neurocognitive disorders.
There are six subtypes of the disease. The behavioral variant (bvFTD) can be
described by noted changes in behavior, personality, and social functioning.
Symptoms may include apathy, lack of empathy, socially inappropriate behavior, and
a decline in personal hygiene. The semantic variant primary progressive aphasia
(svPPA) is a subtype characterized by a progressive decline in language functioning,
including difficulty with word finding, word comprehension, and loss of knowledge of
word meanings. These patients may eventually lose the ability to speak altogether.
Progressive nonfluent variant primary progressive aphasia (PNFA) is a rare form of
FTD that also affects language and speech production. Patients have speech that can be
described as difficult, halting, with grammatical errors and difficulty with word
retrieval. There are three subtypes of FTD that have prominent motor symptoms.
These include corticobasal degeneration (CBD), progressive supranuclear palsy (PSP),
and FTD with motor neuron disease amyotrophic lateral sclerosis (ALS).
Symptoms of FTD are variable and depend on the subtype of the disease but most
show some changes in behavior, personality, language, and movement. Movement
disorders such as Parkinsonism may also occur in some subtypes of FTD. The initial
presentation of FTD is often mistaken for depression and accurate diagnosis requires a
high index of suspicion. A history of insidious onset of apathy and lack of concern for
self should raise the possibility of FTD and we strongly recommend referral to a geriatric
psychiatrist or a neurologist for early accurate diagnosis.
The exact causes of FTD are not yet fully understood, but genetic mutations have
been identified as a significant risk factor. Familial FTD accounts for approximately one-
third to one-half of all FTD cases and presents more commonly as bvFTD than other
FTD subtypes [1]. Mutations in microtubule-associated protein tau (MAPT), progranu-
lin (PGRN), and chromosome 9 open reading frame 72 (C9orf72) expansion mutations
have been found to be the most common causes of familial FTD. In addition, abnormal
protein deposits in the brain (tau, TDP-43, and FUS) have also been implicated in the
development of FTD. The abnormal accumulation of tau and TDP-43 proteins causes
damage to the nerve cells in the brain, leading to cell death and eventually symptoms
of FTD.
Diagnosis of FTD involves a comprehensive neurological evaluation, including labs
and MRI or CT, as well as neuropsychological testing to assess cognitive and language
function. In some cases, genetic testing may also be recommended. Definitive serum or
csf biomarkers for FTD have been elusive.
Possible, probable, and definitive diagnosis of bvFTD can be made using the
following criteria developed by the International Behavioral Variant FTD Criteria
Consortium in 2011. Patients must all show a progressive decline in behavior as observed
by a reliable caregiver.
Possible bvFTD is diagnosed if three or more of the following are present: (a) early
behavioral disinhibition described as socially inappropriate behavior, loss of manners/
decorum, impulsivity, and rash actions; (b) early apathy or inertia; (c) early loss of
empathy or sympathy, including diminished response to other people’s needs or sympa-
thies, and diminished social interest; (d) early perseverative or compulsive/ritualistic
behavior, (i.e. simple repetitive movements, complex, compulsive, or ritualistic behavior,
22 Neurocognitive Disorders
stereotypy of speech); (e) hyperorality and dietary changes, including altered food
preferences, binge eating, increased consumption of alcohol and cigarettes, oral explor-
ation or consumption of inedible objects; (f ) neuropsychological profile demonstrates
deficits in executive functioning and relative sparing of episodic memory and
visuospatial functioning.
Probable bvFTD diagnosis would need to meet the criteria for possible bvFTD, but
also includes imaging with frontal or anterior atrophy on MRI or CT. Alternatively, PET
or single-photon emission computed tomography (SPECT) imaging demonstrating
hypometabolism in the frontal or anterior temporal regions could be used to diagnose
probable bvFTD.
Definitive diagnosis of bvFTD requires the patient to meet the criteria for possible or
probable bvFTD but must also include histopathological evidence of FTD on biopsy or at
post mortem or the presence of a known pathogenic mutation.
The diagnosis of the PPA variant of FTD is made when clinician evaluation shows a
patient to have prominent and progressive difficulty with language as the primary source
of impairment in daily activities. Aphasia not otherwise attributed to another neurop-
sychiatric disorder is the most notable language deficit noted. There are several variants
to the PPA variant of FTD [2].
There is no cure for the disease. Speech and language therapy can be useful in
patients to help them maintain social and language functions and physical therapy can
be used to help maintain motor functions in those with variants that have prominent
motor symptoms. Patient-centered nonpharmacologic interventions focused on
decreasing triggers of problematic behaviors should be attempted prior to or concur-
rent with pharmacologic interventions. There have been studies that have reported
improvements in behavioral symptoms with selective serotonin reuptake inhibitors
and stimulants have been tried with some success to address disinhibition, apathy, and
risk-taking behavior. Studies with memantine and cholinesterase inhibitors have
shown no benefit.
Antipsychotics are options for the treatment of agitation and psychosis but should be
used with extra caution. Riluzole is FDA-approved for the treatment of the ALS variant
of FTD and has been shown to have modestly prolonged survival. Its mechanism is to
reduce the release of glutamate, an excitatory transmitter that is involved in the degener-
ation of motor neurons in ALS.
Take-Home Points
Frontotemporal dementia is the name of a group of dementias that primarily involve the frontal
and temporal lobes of the brain. They are progressive neurodegenerative disorders that are
typically diagnosed in individuals under the age of 65.
Diagnosis of FTD involves a comprehensive neurological evaluation, including labs and MRI or CT,
as well as neuropsychological testing to assess cognitive and language function. In some cases,
genetic testing may be recommended.
Symptoms of FTD are variable and depend on the subtype of disease but most show some changes
in behavior, personality, language, and movement. A history of insidious onset of apathy and lack of
concern for self should raise the possibility of FTD.
There is no cure for the disease. Speech and language therapy can be useful in patients to help
them maintain social and language functions. Physical therapy can be used to help maintain motor
functions in those with variants that have prominent motor symptoms.
“I Am Okay” 23
Further Reading
Desai, A., & Grossberg, G. (2018). Psychiatric
Consultation in Long-Term Care. A Guide
for Healthcare Professionals. 2nd ed.
(Cambridge University Press.)
Section 1 Neurocognitive Disorders
“Leave Me Alone”
Case
6 Agitation in MNCD
For several weeks, Mrs. L, who has MNCD due to probable Alzheimer’s disease in a
moderate stage, had been growing agitated in the afternoon and evening and was
attempting to leave the long-term care facility. She would become physically aggressive
when prevented from leaving and often yelled, “Leave me alone, let me go.”
Mrs. L was taking citalopram 20 mg daily, donepezil 10 mg daily in the morning, and
memantine 10 mg twice daily for MNCD and anxiety symptoms. These medications had
helped decrease Mrs. L’s agitation to some extent, but she continued to have at least one
incident a week of physical aggression toward staff.
Mrs. L was referred to a consultant psychiatrist for potential medication changes to
“control her aggression.” After a comprehensive psychiatric evaluation with team discus-
sion, the psychiatrist ruled out easily correctable causes (e.g. anticholinergic medication,
medical conditions). The psychiatrist met with the team members (Mrs. L’s family, a
certified nursing assistant, licensed practical nurse, and social worker) and discussed various
potential SPPEICE that could be instituted. Mrs. L’s daughter mentioned that her mother
had always loved to work in the house rather than watch TV or play games. Mrs. L had never
had pets and did not like them and had never been a regular listener of music.
The team decided to give Mrs. L simple tasks, such as setting the table, folding
napkins, and putting letters in envelopes. Other interventions, such as pets or music
groups, were not selected. After two weeks of implementing these person-centered
approaches, Mrs. L had become significantly less distressed, as shown by a reduction
in agitated behaviors, and was more easily reassured and redirected.
Teaching Points
Agitation is a neuropsychiatric syndrome that is commonly seen in those with MNCDs.
Those demonstrating agitation can show an increase in motor activity, restlessness,
emotional distress, and physical or verbal aggressiveness. The estimated prevalence of
agitation in those with MNCD is estimated to be 30–50% of those with Alzheimer’s
disease, 30% of those with DLB, 40% in FTD, and 40% in vascular dementia. It is the
third most common neuropsychiatric symptom in dementia after apathy and depression.
Up to 80% of people with dementia experience some degree of agitation at some point
during the course of their illness.
Agitation in dementia can take many forms such as restlessness, pacing, verbal or
physical aggression, and resistance to care. It can be triggered by a variety of factors,
including pain, discomfort, environmental changes, and communication difficulties.
24
“Leave Me Alone” 25
Collectively, agitation in those with MNCD produces a very high burden to the health-
care system. Agitation increases caregiver burnout, the need for hospital admissions, and
medication burden, and even increases mortality.
Assessing agitation can be somewhat subjective but this can be aided by the use of
rating scales such as the Pittsburgh agitation scale [1]. The Cohen-Mansfield Agitation
Inventory is a rating scale filled out by caregivers that can help assess the degree of
agitation and response to treatment [2].
There are several possible overlapping causes or triggers of agitation in MNCD.
Agitation is likely related to degenerative changes in the brain that regulate mood and
behavior, more specifically, pathology in the function of the anterior cingulate cortex and
orbitofrontal cortex. Increased sensitivity to noradrenergic signaling has been described,
which is possibly due to a frontal lobe upregulation of adrenergic receptors that happens
as a reaction to the depletion of noradrenergic neurons within the locus coeruleus. This
might explain the abnormal reactivity to weak stimuli and global arousal found in many
patients with MNCDs.
Agitation can be a normal response for those with MNCD when they are experi-
encing physical discomfort or pain and seemingly innocuous changes in the environ-
ment can also be triggers. Examples include changes in routine, moving to a new room
or facility, surroundings that are too warm or cool, hunger, or an increase in ambient
noise. Agitation can also be seen in dysfunction in social interactions such as those seen
with caregiver stress and loneliness.
Prevention of agitation is the goal. Knowing a resident well includes knowing a
resident’s personality, favorite people, preferences for activities, preferences for foods,
personal and family history, and a wide variety of other factors. Long-term care facilities
often make a point of including pages of intake information when admitting a new
resident in an effort to provide a more comfortable environment for them.
In regard to the case of Mrs. L, attempts were made to get to know the patient better.
Knowing the resident well (including the resident’s role in life before entering long-term
care, the resident’s daily interests, and what activities give the resident meaning in life
[life history]) is key to significantly improving the success rate of SPPEICE. If the team
had not tried to address Mrs. L’s underlying need to feel useful and engage in activities
that gave meaning to her life, her agitation and aggression could have continued and she
may even have been prescribed antipsychotic medication, putting her at risk of acceler-
ated cognitive and functional decline.
It is good to note that nonpharmacologic interventions are often key to minimizing
episodes of agitation in those with MNCD. Environmental interventions focus on
changing the living environment to reduce agitating triggers and create a calmer
atmosphere. Reducing ambient noise levels, providing adequate lighting, ensuring a
comfortable temperature, and trying to create a home-like environment as much as
possible can be helpful environmental interventions.
Sensory interventions focus on reducing sensory inputs that can trigger agitation and
increasing sensory inputs that can improve relaxation. Making sure that residents have a
favorite comfortable chair to sit in and a comfortable warm bed to sleep in is key. Shower
times can be easier when a resident is not exposed to water that they may find too hot or
cold. Washing one body area at a time while others are kept covered can also lead to
more comfort and dignity. Living areas that are quieter and more private can allow
residents to have some quiet time to themselves. Patient-tailored sensory interventions
26 Neurocognitive Disorders
that can be helpful also include massage therapy, music therapy, aromatherapy, and
therapeutic touch. The utilization of music in long-term care settings has received
particularly favorable attention in the literature [3].
Psychosocial interventions focus on addressing the underlying psychological and
emotional needs of the patient that may contribute to agitation. Interventions can
include providing social interactions with other residents such as “mocktail” hour,
games, and group entertainment. Some patients may respond positively to being
involved in simple “chores” that can help them feel more useful and less bored. Pet
therapy is becoming more common in long-term care settings and can provide emo-
tional comfort and a way to connect with others. Exercise programs not only provide an
opportunity for maintaining strength and agility but can also provide another venue for
interacting with peers.
Social interventions should be tailored to the individual needs and preferences of the
patient. What works for one patient may not work for another, so it is important to
involve the patient and their caregivers in the intervention planning process.
Additionally, social interventions should be implemented in conjunction with other
treatments, such as medication or medical interventions, as appropriate.
The pharmacologic management of agitation in those with MNCDs is complex
and many studies have shed light on the topic. The treatment may depend on the
acuity of the agitated behaviors and the underlying subtype of MNCD. In those with
Alzheimer’s disease, selective serotonin reuptake inhibitors (SSRIs), trazodone, mir-
tazapine, prazosin, and low-dose atypical antipsychotics can be trialed. At the time
of writing, brexpiprazole had recently been FDA approved for the treatment of
agitation in Alzheimer’s disease [4]. Rivastigmine can be trialed in those with DLB
or PD-D [5].
Agitation can also be related to dopaminergic drugs such as carbidopa/levodopa,
particularly when combined with SSRIs or serotonin–norepinephrine reuptake inhibi-
tors (SNRIs). If an atypical antipsychotic is thought to be warranted, an agent with low
D2 receptor affinity such as quetiapine is probably preferred. Trazodone, SSRIs, and
atypical antipsychotics can be tried for patients with agitation related to frontotemporal
dementia. Those with vascular dementia may respond well to SSRI with second- or third-
generation antipsychotics being preferred in more significant agitation or agitation with
psychosis.
Take-Home Points
Agitation is a neuropsychiatric syndrome that is commonly seen in those with MNCDs. Those
demonstrating agitation can show an increase in motor activity, restlessness, emotional distress,
and physical or verbal aggressiveness.
Agitation in dementia can take many forms such as restlessness, pacing, verbal or physical
aggression, and resistance to care.
Agitation is the third most common neuropsychiatric symptom in dementia after apathy and
depression. Up to 80% of people with dementia experience some degree of agitation at some
point during the course of their illness.
The pharmacologic management of agitation in those with MNCDs is complex and many studies
have shed light on the topic. At the time of writing brexpiprazole had recently been FDA approved
for the treatment of agitation in Alzheimer’s disease.
“Leave Me Alone” 27
“I Am Fine”
Case
7 Apathy Syndrome
Mr. L’s successful professional career. The psychiatrist counseled Mrs. L not to insist
that Mr. L attend social and family events and to lower her expectations regarding
daily bathing, becoming content with twice-weekly bathing. The psychiatrist also
recommended that she see a social worker for individual counseling to address the
stress of caregiving and the grief of losing parts of her husband’s original personality.
After 12 weeks, Mrs. L’s depression had decreased significantly, and Mr. L showed
mild interest in music therapy and in his life history project. He was agreeable to
continuing these activities.
Teaching Points
Apathy syndrome is a frequently observed condition among older adults, particularly in
long-term care environments, and its prevalence varies depending on the specific
population under study. Estimates suggest that up to 70% of individuals with
Alzheimer’s disease, 40% of those with Parkinson’s disease, and 38% of those with late-
life depression may experience symptoms of apathy [1]. It is also commonly seen in
those with a history of TBI and stroke but can be seen in virtually any neurodegenerative
disease.
Apathy syndrome is a clinical condition characterized by a lack of motivation,
interest, or emotional responsiveness. It involves a reduced or diminished ability to
initiate and sustain goal-directed behavior, leading to a general indifference or disin-
terest in one’s surroundings, activities, or social interactions. Individuals with apathy
syndrome may exhibit a lack of enthusiasm, initiative, or drive, often appearing
uninterested or detached from their usual activities or responsibilities. They may
display decreased involvement in hobbies, reduced social interaction, and diminished
emotional expression. Apathy syndrome can significantly impact a person’s daily
functioning, productivity, and overall quality of life. Patients do not meet the criteria
for apathy if symptoms can be better explained by other factors such as intellectual,
physical, or motor disability, changes in level of consciousness, or the direct effect of
substance use.
Depression is also common in neurocognitive disorders, making the need to distin-
guish between the two conditions more pertinent for those who regularly care for those in
the long-term care setting. It is possible to have comorbid depression and apathy syndrome
associated with the underlying neurocognitive disease. Co-occurrence rates of apathy and
depression are estimated at 14–38% in those with neurocognitive disorders [2].
The Apathy Evaluation Scale was developed by Robert S. Marin, MD, and has several
versions that can be self-administered or administered by a caregiver or clinician [3]. The
problem does not come from the recognition of apathy syndrome, but in the treatment.
Medical treatment should focus on optimizing treatment of the underlying condition,
such as Alzheimer’s or Parkinson’s disease, or underlying neurocognitive disorder.
Attention should be paid to rule out underlying psychiatric conditions such as depres-
sion. Apathy is common in depression, but straightforward apathy syndrome lacks the
typical symptoms of depression such as sadness, helplessness, hopelessness, impaired
sleep, changes in appetite, and so on.
Apathy is often most concerning to family members. Concerned caregivers will most
often bring the symptoms of apathy to the attention of the clinician. Apathy is associated
with a high degree of caregiver burden and distress.
30 Neurocognitive Disorders
Box 7.1 Suggested approach to starting an older adult patient with apathy syndrome
on methylphenidate
1. Identify a patient with potential apathy syndrome.
2. Rule out potential medical mimickers of apathy syndrome such as hypothyroidism,
medication side effects, and depression.
3. Institute some nonpharmacologic interventions as listed in Table 2.
4. Take extra caution and gain informed consent from patients with apathy syndrome, a
history of stroke, coronary artery disease, or tachyarrhythmias. Stimulants have shown
an increased risk of adverse cerebrovascular and cardiac complications in older adults
in the short term. These negative effects may attenuate with time.
5. Discuss the use of stimulant medication with the patient and surrogate. Explain the
risks and benefits of the medication.
6. Initiate methylphenidate at 5 mg twice daily (morning and midday). Evaluate for effect
and tolerability after 2–4 weeks.
7. Consider increasing the dose of methylphenidate to 10 mg twice daily if inadequate
response to 5 mg bid dosing.
8. Re-evaluate after another 4–6 weeks. Consider discontinuing medication if no
improvement in apathy syndrome is noted with methylphenidate 10 mg bid dosing.
“I Am Fine” 31
Box 7.2 Nonpharmacologic interventions that may help with apathy syndrome
1. Consider being more authoritarian by initiating potentially pleasurable activities
for those with apathy syndrome. For example, a statement such as, “It’s time
for our walk,” may be better received than, “Would you like to go for a
walk now?”
2. Establish routines for scheduled activities. A schedule that includes regular activities
such as exercise, crafts, socializing, games, and outdoor activities can create positive
momentum and help to combat apathy.
3. Make sure activities are appropriate to the level of function for those with minor or
major neurocognitive disease. A short game of Yahtzee or bingo may be more well-
received than an outing to the symphony, for example.
4. Gradually increase the level of engagement. Start with short, easy, pleasurable
activities and gradually work up to those activities that may be more involved.
5. Create a living environment that is full of stimulating and interesting things.
6. Tie together activities that are less appealing to a patient with activities that are always
appealing to a patient. For example, stopping for an ice cream cone after a visit to the
pharmacy or a trip to the doctor’s office.
7. Consider including exercise, music therapy, multisensory stimulation, and pet therapy
in the regular schedule of a person with apathy syndrome.
Take-Home Points
Ms. L was an 84-year-old widow who had MNCD due to probable Alzheimer’s disease
and was admitted to an assisted living home because of increasing problems with
agitation, incontinence, lack of self-care, and wandering. She had been in the AL home
for more than six months. The family had noticed that, even when Ms. L had been living
in her home, she talked about children coming into her house and bringing cats and
dogs. Over four months, Ms. L was having periods of agitation and anxiety over “little
children” going through her belongings. She was also complaining of being upset with
the AL staff because they would allow cats and dogs to roam freely into her room. Ms.
L was referred to the psychiatrist for the management of hallucinations.
On evaluation, she was not taking any medication that could have caused hallucin-
ations, nor was she having any physical aggression or depressive symptoms. Ms. L was
taking memantine 10 mg twice daily and donepezil 10 mg daily, as well as medications
for hypertension (hydrochlorothiazide and atenolol) and hyperlipidemia (simvastatin).
She did not complain of any new physical health problems, and family and staff reported
that episodes of agitation and hallucination occurred once or twice a week. Staff reported
that Ms. L participated in activities, slept well, and had a good appetite.
When the psychiatrist inquired how staff and family dealt with Ms. L’s complaints,
they reported that they would try to convince Ms. L that she was imagining things and
that there were no children or animals coming to her room. Staff and family said that
often this would agitate Ms. L even more. The psychiatrist inquired about Ms. L’s
personality before the MNCD, and the family reported that she had always been active
and liked to be busy and “on the go.”
During the interview, Ms. L denied having any problems, denied being in physical
pain, reported feeling “fine,” said “yes” to feeling bored and lonely, but reported good
sleep and appetite. She stated that “those children are cute” and did not seem bothered by
their presence in her room. Her mental status exam showed significant short-term
memory loss (recall was 0/3), and her SLUMS score was 12.
The psychiatrist explained to the family and staff that visual hallucinations were a
symptom of MNCD such as Alzheimer’s disease and that current symptoms were best
managed with SPPEICE [1]. The psychiatrist recommended that family and staff not
correct or argue with Ms. L, but try to reassure and then distract her. The psychiatrist,
together with family and staff, devised a daily activity schedule that would involve more
activities that staff and family could engage Ms. L in (e.g. reminiscing with old photos,
listening to preferred music, reading the Bible to Ms. L) and a short nap in the afternoon.
Ms. L continued to have transient visual hallucinations but her agitation during those
33
34 Neurocognitive Disorders
periods gradually decreased, and the staff became more adept at distracting Ms. L after
reassuring her that they would “take care” of the problem promptly.
Teaching Points
Alzheimer’s disease is the most common form of MNCD, causing gradual declines in
many areas of cognition. Alzheimer’s disease accounts for 60–70% of cases of dementia
worldwide and causes a gradual deterioration of various cognitive domains, including
decision-making, language, memory, learning, orientation, and judgment [2]. The exact
pathophysiology of cognitive decline in dementia is still being intensely studied but one
of the pillars of our understanding of Alzheimer’s disease and some other dementias is a
gradual development of acetylcholine deficiency in key neural pathways. Cholinesterase
inhibitors such as donepezil, rivastigmine, and galantamine slow down the breakdown of
acetylcholine within the synapses, thereby effectively increasing the available acetylchol-
ine within neural pathways involved in cognition (Table 8.1).
Donepezil is a prototypical cholinesterase inhibitor (ChEi) and was approved for use
in 1996. It is FDA-approved for the treatment of mild, moderate, or severe Alzheimer’s
disease and is considered a symptomatic treatment for Alzheimer’s disease and some
other dementias as it does little to alter the course of the disease. It may have benefits in
cognition, mild neuropsychiatric symptoms, and possibly delay the need for transfer to a
Table 8.1 Medications commonly used in MNCD: Cholinesterase inhibitors and memantine.
Medication Starting Dose and Dosing Schedule Therapeutic Daily Dose (Range)
Donepezil 5 mg once a day after meals, may 5–23 mg
increase every four weeks by 5 mg to
highest tolerated therapeutic daily
dose
Rivastigmine 4.6 mg/24-hour patch applied on a 9.5–13.3 mg
transdermal clean, dry, hairless part (upper back
patch usually), may be increased every four
weeks to the highest tolerated
therapeutic daily dose
Rivastigmine 1.5 mg twice daily after meals, may 6–12 mg
increase every four weeks by 3 mg/day
to the highest tolerated therapeutic
daily dose
Galantamine 8 mg once daily after meals, may 16–24 mg
extended increase every four weeks by 8 mg to
release the highest tolerated therapeutic daily
dose
Memantine 7 mg once daily, may increase by 7 mg 14–28 mg (7–14 mg for residents
extended every seven days to the highest who have creatine clearance less
release tolerated therapeutic daily dose than 30)
Memantine 5 mg once a day, may increase by 20 mg in two divided doses
5 mg per week to the highest (5–10 mg for residents who have
tolerated therapeutic daily dose creatine clearance less than 30)
“Those Children Are Cute” 35
care facility for those with mild-to-moderate disease. The side effects of donepezil and
other cholinesterase inhibitors are well-known and include possible insomnia, diarrhea,
muscle cramps, nausea, and loss of appetite. Donepezil is usually dosed in the morning
due to the risk of insomnia and should be avoided, along with other cholinesterase
inhibitors, in those with known bradyarrhythmia or sick sinus syndrome.
When counseling patients and families on the use of cholinesterase inhibitors it is
important to inform them that studies of these medications indicate that improvements
in measures of cognition are generally modest. Side effects are also somewhat common
and are usually GI-related. A trial of donepezil is warranted for nearly all patients with
Alzheimer’s disease. Rivastigmine is FDA-approved for Parkinson’s dementia and is
often used off-label for DLB. There is less support for the use of ChEis in vascular
dementia and they are not indicated for FTD.
Dosing of donepezil usually starts at 5 mg daily. If a patient or caregiver notes improve-
ment in cognition, it is appropriate to encourage the patient to continue with the medication
and increase the dose to 10 mg daily if tolerated. If there are GI side effects to lower doses of
donepezil, a few different approaches can be taken. A dose of donepezil can be taken at
bedtime, which may help with the experience of GI side effects. Donepezil can also be
switched to transdermal rivastigmine. Transdermal rivastigmine, although more costly, is
known to have much lower GI side effects. A more complete list of strategies to deal with
ChEi intolerance is presented in Box 8.1. If these strategies to improve tolerance of the
medication are unsuccessful or there are other side effects or worsening of neuropsychiatric
symptoms it is probably advisable to discontinue the medication.
Another medication that is FDA-approved for the treatment of moderate-to-severe
Alzheimer’s disease is memantine. It acts as an NMDA receptor antagonist within the
CNS and thereby helps to blunt the excitotoxic effects of glutamate and potentially delay
the progression of the disease. Memantine is generally well tolerated when titrated to a
goal dose of 20 mg daily. Some patients may experience an exacerbation of neuropsy-
chiatric symptoms with memantine, and this may be a reason for discontinuing the
medication during the titration phase. Memantine is probably more effective as a
disease-modifying agent than as a medication to improve symptoms of MNCD [3].
It can be used either alone or in combination with acetylcholinesterase inhibitors.
The use of a combination of cholinesterase inhibitor and memantine is recommended
for all Alzheimer’s disease patients if tolerated [4]. In December 2014, the FDA approved the
combination of donepezil and memantine extended release for patients with moderate-to-
severe Alzheimer’s disease. Combination therapy has been shown to provide benefits over
ChEis alone and may postpone the need for admission to a skilled nursing facility. The
increased cost of the fixed-dose combination donepezil/memantine formulation should be
weighed against the lower cost of standard generic donepezil/memantine separate product
dosing. The fixed-dose combination formulation may be useful for those who desire
decreased pill burden and those with dysphagia as it can easily be sprinkled onto soft foods.
Once a ChEi has been started and seems well tolerated it may be a challenge to keep
patients and caregivers motivated to continue the medication, especially as major
neurocognitive disorders will invariably continue to produce cognitive and behavioral
declines. In these cases, it can be helpful to remind caregivers that there are benefits to
continuing medications for dementia even in light of continued decline. The advantages
to maintaining the medication include continued slowing of the decline in cognition,
delaying the need for a higher level of care, and potentially delaying the need for
medications to address neuropsychiatric symptoms.
Another scenario to occasionally present itself to a provider who often cares for older
adults is when to stop medications for dementia when the dementia is quite advanced.
The stages of MNCD often progress in a stepwise fashion according to the Functional
Assessment Staging (FAST) scale (see Figure 8.1). The FAST scale was developed by
Dr. Barry Reisberg and his colleagues at the New York University School of Medicine’s
Aging and Dementia Research Center. The scale was designed to assess and stage the
functional decline associated with Alzheimer’s disease and other dementias. It provides a
framework for tracking and describing the progression of functional impairment in
individuals with dementia.
Those patients with FAST scores in the 7s are considered to be in the final stage of
the disease. During this stage, there may be a question as to the continued benefit of
dementia medications. Caregivers should be given enough information to make an
informed shared decision with the clinician. On one hand, discontinuing ChEis in
those who have taken them for some time can potentially lead to decreased cognitive
function or an increase in neuropsychiatric symptoms. On the other hand, discon-
tinuing these medications could lead to a decreased pill burden and potentially
decreased side effects of the medication(s). It can be difficult for those with advanced
dementia to voice concern for side effects and it can be reasonable to discontinue
ChEis in those with suspicion of GI side effects (decreased appetite, nausea/vomiting,
diarrhea, signs of abdominal discomfort). Questions to consider with caregivers when
deciding when to discontinue medications for dementia are presented more fully in
Box 8.2.
Take-Home Points
Alzheimer’s disease is the most common form of MNCD, causing a gradual decline in many areas of
cognition. Alzheimer’s disease accounts for 60–70% of cases of dementia worldwide.
Once a ChEi has been started and seems well tolerated it may be a challenge to keep patients and
caregivers motivated to continue the medication, especially as MNCDs will invariably continue to
produce cognitive and behavioral declines.
Donepezil is a prototypical ChEi and was approved for use in 1996. It is FDA-approved for the
treatment of mild, moderate, or severe Alzheimer’s disease and is considered a symptomatic
treatment for Alzheimer’s disease and some other dementias as it does little to alter the course
of the disease.
The advantages of maintaining patients on ChEis include continued slowing of the decline in
cognition, delaying the need for a higher level of care, and potentially delaying the need for
medications to address neuropsychiatric symptoms.
38 Neurocognitive Disorders
9 Agitation in MNCD
Mrs. S was a 78-year-old woman who had DLB and was living in an AL home. Over
several months, she had been expressing concerns that men were in her “home” telling
her to do inappropriate acts like take down her pants. Mrs. S would typically start yelling
at 3 pm that she can see “the man” in the trees looking into her room. She was not
sleeping well, and she used foul language toward staff during personal care. Over a
period of four weeks, Mrs. S had become physically aggressive toward staff, hitting,
slapping, kicking, and biting them. Individualized activities and attention had
initially been successful, but during those four weeks her behavior was not manageable.
She was referred to the consulting psychiatrist for the management of aggression.
On examination, the psychiatrist found that Mrs. S had mild muscle stiffness, restricted
affect, and persecutory delusions. The medication review did not identify any medication
as potentially being responsible. She was taking rivastigmine 3 mg twice daily with meals.
She had a history of not tolerating a higher dose of rivastigmine and could not tolerate
memantine. Laboratory tests were within normal limits and urine analysis was negative
for infection.
Staff and family were counseled regarding the possibility of engaging in personalized
activities and interventions to reduce Mrs. S’s agitation. For example, one staff member
would engage Mrs. S from 2 pm onward in a coloring activity and another staff member
would close the curtains in her room so that when Mrs. S would go there she would not
see trees outside her window. Additionally, only female staff were allowed to provide
personal care.
The risk of stroke and premature death associated with the use of atypical antipsy-
chotics was explained to the family, as well as the risks of not giving an antipsychotic at
all. The family agreed to a trial of low-dose quetiapine. Mrs. S was started on quetiapine
12.5 mg in the morning and at bedtime daily. She developed daytime sedation and hence
her quetiapine was shifted to 25 mg daily at bedtime. After one week, her sleep had
improved but the agitation and aggression persisted, so the quetiapine was increased to
37.5 mg daily at bedtime and, after four days, to 50 mg daily at bedtime. She had mild
daytime sedation, but it resolved over five days. Over the next two weeks, her agitation
and aggression improved significantly, she allowed most activities of daily living (ADL)
care, and stated, “I am so glad those terrible men have left,” and felt relieved. She still
expressed periodic concern that the men were coming and going through her suitcases
and her drawers but could be reassured and distracted most of the time. After three
months, the quetiapine was tapered and then discontinued over four weeks. Mrs. S did
not show any worsening symptoms.
39
40 Neurocognitive Disorders
Teaching Points
This case highlights an important topic in the treatment of those with MNCD. Those
with MNCD can demonstrate a wide variety of behaviors that may be considered
disruptive and can be attributed to a patient’s underlying neuropathology. Terms for
the behavioral disturbances seen in MNCD have been referred to a number of ways,
including behavioral disturbances, behavioral and psychological symptoms of dementia,
responsive behaviors, biopsychosocial–spiritual distress (BPSD), and simple agitation.
The term BPSD is the term this author prefers as it stresses that behavioral issues in
MNCD are often contributed to by a patient’s unmet needs.
The biological and psychological/emotional needs of those with MNCD are the same
as those without. The needs of all persons were presented as a “hierarchy of needs” by
Abraham Maslow in 1943 and his model is still a useful way of conceptualizing human
needs today. Physiological or biological needs are considered the most basic and essential
for life and include needs such as air, food, water, shelter, and sleep. Safety needs come
next in importance and involve being in a safe, secure environment, free from the
potential of harm. Needs for love and belonging come next and include the needs of a
person to have friendships, love, intimacy, and social connectedness. Esteem needs are
one step further up the pyramid and include maintaining dignity and having the respect
of others. Self-actualization needs include a person’s being able to grow and live up to
one’s full potential. See Box 9.1.
Biopsychosocial–spiritual distress can be experienced by anyone with unmet needs in
any of the above areas. Those with MNCD may be unable to verbally describe their
unmet needs but often give other clues as to their presence. Common behavioral signs of
unmet biopsychosocial needs are shown in Box 9.2.
Identifying the behaviors of BPSD is the easiest part of the process of addressing
them. These behaviors are usually first noted by caregivers or the family of the patient.
The clinician should resist the urge to quickly turn to pharmacologic interventions for
2. Safety needs.
Having a safe and secure environment free from the threat of physical or emotional
harm. Being free of pain.
4. Esteem needs.
Maintaining dignity and having the respect of others.
5. Self-actualization needs.
Being able to live up to one’s full potential.
“Those Terrible Men Have Left” 41
Box 9.3 Potential unmet biopsychosocial needs in the long-term care setting
1. Loneliness. Is the resident lacking in meaningful connection with loved ones? Has there
been a recent loss of a loved one? Potential interventions: Calling involved family
members and encouraging visits. Is there a volunteer program to provide visits to
those with MNCD? Is pet therapy available? Consider doll therapy for those with
moderate–severe MNCD.
2. Boredom. Is there a lack of appropriate activities for the patient? Potential
interventions: Ask the activities director and care staff to encourage the patient to
participate in scheduled activities or explore new activities. Ask the family about the
patient’s interests prior to entering the long-term care facility.
3. Pain. Ask about any suspicion of a recent fall or injury, undertreated musculoskeletal
pain, urinary tract infection, skin breakdown, or history of headaches, and so on.
Potential interventions: Trial of scheduled acetaminophen to treat chronic pain. Trial of
duloxetine for the treatment of chronic pain.
4. Hunger. Ask about eating habits. Is the patient eating a light meal or missing meals and
becoming agitated prior to the next meal? Potential intervention: Offer appealing
snacks in between meals.
5. Toileting issues. Is the resident wet due to infrequent changes? Is the resident guided
to the bathroom on a regular basis for bowel movements? Is constipation possible?
6. Difficulty communicating needs. Those with MNCD may have difficulty expressing their
needs and wants.
7 Medical interventions: Does the resident have any treatments that are potentially
uncomfortable? Is there any room to deprescribe any potentially unnecessary
medications that may be contributing to the cumulative side effect burden?
8. Changes in routine. Have there been any potentially anxiety-provoking changes in day-
to-day routines?
9. Interpersonal conflicts. Are there any new relationships that could be anxiety-
provoking to the resident? Sometimes the loss of a trusted caregiver or the
introduction of an unfamiliar one can provoke signs of BPSD.
10. Staff issues. Are staff members overworked? Those with MNCD can sense the distress
of others and may react in a like fashion.
11. Issues with other residents. Are there other residents with behavioral issues that may
be creating a threatened sense of safety and security?
that are experiencing psychosis. Antipsychotic use should be limited due to the well-
known black box status of these medications. It should also be kept in mind that
psychosis may be more subtly demonstrated in those with MNCD and thereby more
difficult to detect. These patients may not be able to describe their hallucinations or be
willing to freely discuss delusions. Some more subtle signs of psychosis in those with
MNCD are listed in Box 9.5.
The choice of antipsychotics in older adults with agitation and psychosis is a
potentially complicated topic. A clinician would be well served by becoming comfortable
with the use of one or two antipsychotics. Every attempt should be made to avoid first-
generation antipsychotics as newer atypical antipsychotics are generally better tolerated.
At the time of writing, the medication brexpiprazole had recently been FDA-approved
for the treatment of agitation in Alzheimer’s disease. Barriers to the use of this medica-
tion will undoubtedly be cost-related due to the medication’s current brand name-
only status.
“Those Terrible Men Have Left” 43
Box 9.4 Potential therapeutic options for anxiety and agitation in those with MNCD
Consider:
1. SSRIs (sertraline, fluoxetine). Can be quite effective if suspicion of an underlying mood
disorder. May take weeks for clinical response.
2. SNRIs (duloxetine/venlafaxine). Consider if underlying chronic pain. May take weeks for
clinical response. Tapering can cause mild to moderately uncomfortable
withdrawal syndrome.
3. Trazodone. Generally considered safe in older adults. Mild sedating effects of this
antidepressant can be effective for anxiety. Results can be seen soon after the dose is
given. Consider routine administration if there are regular signs and symptoms of
anxiety. Can be given prn.
4. Buspirone. Can be effective. Often prescribed at an inadequate dose. The dose should
be increased to at least 30 mg daily (divided). May take some weeks for a response.
5. Gabapentin. The anxiolytic effects can be brisk. Generally well tolerated at low to
moderate doses. Avoid high doses in older adults due to side effects. Must be dosed
2–3 times daily due to short half-life.
6. Mirtazapine. Low doses of 7.5–15 mg have a mild sedative effect. Dosed at night. May
help with nighttime anxiety and insomnia. Good appetite stimulant and may be a good
choice if anxiety with comorbid malnutrition.
7. Propranolol. Small doses of propranolol (10 mg bid) can be helpful.
Avoid:
1. Benzodiazepines. Older adults can have worsening cognition and worsening
disinhibited behaviors and increased falls with benzodiazepines. Risks usually
outweigh benefits.
2. Antihistamines (hydroxyzine, diphenhydramine). Although well tolerated and effective
in younger adults they should be avoided in older adults due to anticholinergic effects.
3. Antipsychotics (quetiapine, risperidone, olanzapine, aripiprazole). These should be
reserved for those who may be experiencing psychotic symptoms.
If antipsychotics are indicated, they must be prescribed “off label” (all but brexpi-
prazole). For this reason, the provider should clearly document the indication of the need
for the antipsychotic. The suspicion or observations of psychotic behaviors should be
documented in the medical record as it should be clear to any interested party why these
potentially high-risk medications were needed.
44 Neurocognitive Disorders
Special note should also be made when considering antipsychotic choices in those
with MNCD due to Parkinson’s disease or related DLB. These patients are almost
universally sensitive to the D2-blocking effects of nearly all antipsychotics. The best
medications for psychosis in these conditions are clozapine (should probably be pre-
scribed after consulting with a psychiatrist), quetiapine (due to lower affinity for D2
receptors/lower side effects), and pimavanserin (does not bind to D2 receptors).
Pimavanserin is an excellent choice but can be limited by cost and insurance coverage.
It is great when a course of antipsychotic medication is successful in controlling
anxiety or agitation related to psychosis in those with MNCD. Within a relatively brief
period (weeks or months) the provider should consider tapering the medication off or to
the lowest effective dose possible. Although these medications can be quite effective, the
side effects of increased risk of falls and potentially increased cardiovascular compli-
cations warrant limiting their use to the lowest dose possible for the shortest length of
time clinically indicated.
Take-Home Points
Published 2021 Apr 16. doi:10.3389/ Marcinkowska, M., Śniecikowska, J., Fajkis, N.,
fneur.2021.644317 Paśko, P., Franczyk, W., & Kołaczkowski,
Grossberg, G. T., Kohegyi, E., Mergel, V., M. (2020). Management of dementia-
Josiassen, M. K., Meulien, D., Hobart, M., related psychosis, agitation and aggression:
Slomkowski, M., Baker, R. A., McQuade, R. A review of the pharmacology and clinical
D., & Cummings, J. L. (2020). Efficacy and effects of potential drug candidates. CNS
safety of brexpiprazole for the treatment of drugs, 34 (3), 243–268. https://doi.org/10
agitation in Alzheimer’s dementia: Two 12- .1007/s40263–020-00707-7
week, randomized, double-blind, placebo- Watt, J. A., Goodarzi, Z., Veroniki, A. A.,
controlled trials. The American Journal of Nincic, V., Khan, P. A., Ghassemi, M., Lai,
Geriatric Psychiatry, 28 (4), 383–400. Y., Treister, V., Thompson, Y., Schneider,
https://doi.org/10.1016/j.jagp.2019.09.009 R., Tricco, A. C., & Straus, S. E. (2020).
Ijaopo, E. O. (2017). Dementia-related Comparative efficacy of interventions for
agitation: A review of non-pharmacological reducing symptoms of depression in
interventions and analysis of risks and people with dementia: Systematic
benefits of pharmacotherapy. Translational review and network meta-analysis.
Psychiatry, 7 (10), e1250. https://doi.org/10 The BMJ, 372. https://doi.org/10.1136/bmj
.1038/tp.2017.199 .n532
Section 1 Neurocognitive Disorders
Mr. U, a 78-year-old male in the late stages of MNCD due to Alzheimer’s disease, had
developed acute bleeding from the rectum and his wife was not sure if she should admit
him to the hospital or opt for hospice care. The long-term care provider involved in
Mr. U’s treatment helped Mrs. U understand the futility of hospitalization and that
Mr. U would have wanted only comfort care at that point. Mrs. U agreed that her
husband would have wanted comfort care only, but she wanted to be sure that she was
not “giving up” on him. The couple’s daughter and son felt sure that their father would
want comfort care only. After their children’s input, Mrs. U decided to forgo hospital-
ization and opted for hospice care. Her dilemma was understandable because just three
months before, Mr. U was walking, seemed to enjoy his wife’s company, and could feed
himself with assistance although he could not recognize family members (including his
wife) by name. He had developed pneumonia and Mrs. U had opted for hospitalization
and treatment at that time. Although the pneumonia was treated successfully, Mr. U
developed severe agitation and delirium in the hospital and needed antipsychotic medi-
cation to control agitation. When he returned to the nursing home, he used a wheelchair,
had lost the ability to feed himself, had severe speech problems (was extremely difficult
to understand), and was able to articulate only some words but not full sentences.
After Mr. U was enrolled in a hospice, the family and staff were surprised to learn
that two staff members on different occasions had heard Mr. U clearly state, “I saw the
heavenly gates” and “I was told to come back later.” Mrs. U asked their pastor to come
for spiritual support and to help her understand Mr. U’s statements. The pastor
suggested that maybe Mr. U was waiting for his wife to say goodbye and say that it
was okay for him to go. Mrs. U followed the pastor’s advice, and three days later
Mr. U died peacefully with his wife holding his hand and their children present at the
bedside.
Teaching Points
Even in the terminal stages of MNCD, an individual may be able to say complete
sentences on rare occasions. Spiritual needs are more important than ever in the care
of a resident during this time. The help of chaplains, members of the clergy, and fellow
members of the resident’s faith community is crucial for the well-being of many
residents and their families. Families may also need guidance to say “goodbye” and
understand that allowing nature to take its course (and thus allowing a natural dying
process) is not “giving up” but “letting go.”
46
“I Saw the Heavenly Gates” 47
Primary healthcare providers and mental health professionals are likely to be engaged
with family or care staff with questions regarding end-of-life care issues in those in the
terminal stages of MNCDs. Those who provide care in the long-term care setting should
be familiar with issues regarding end-of-life care in MNCD. Medical mental health
providers and primary providers should also be comfortable initiating conversations
with family members of those with progressing MNCDs. Important issues to discuss
with families of those with terminal MNCD are listed in Box 10.1.
Family members may be relatively uninformed when it comes to the true diagnosis of
a declining loved one with MNCD. Systems of care are often fragmented at the end of
life, without the long-term support of a single primary care physician. This fragmenta-
tion of care often leaves families with a suboptimal understanding of diagnoses and
anticipated progression of illness.
Goals of care conversations are often difficult and time-consuming for providers.
This can result in families that may be underprepared for end-of-life issues when they
arise. A suggested template for goals of care conversations is presented in Box 10.2.
Physicians and advanced practice providers (APPs) working with older adults may
receive less training in issues of palliative care and hospice care in those with MNCD
than is ideal. Palliative care is an approach to medical care that focuses on providing
relief from pain, other symptoms, and the physical, emotional, and psychosocial stress
that individuals with serious illnesses may experience. The primary goal of palliative care
is to improve the quality of life for patients and their families, particularly when dealing
with complex, chronic, or terminal medical conditions. Palliative care is usually provided
with the oversight of a physician or APP with additional training in the management of
Box 10.1 Important topics to cover with families of those with late-stage MNCD
1. Diagnosis.
2. Prognosis.
3. Goals of care.
4. Palliative care versus hospice care.
5. Eligibility for hospice care for those with MNCD.
6. Code status.
7. Receiving appropriate legal counsel regarding end-of-life financial/legal matters.
8. Issues involving nutrition and hydration.
Box 10.2 Important topics to discuss when addressing goals of care with patients
and families
1. Assess the level to which patients or surrogates have previously discussed goals
of care.
2. Gauge the level of understanding of patients/families of diagnosis and likely
disease progression.
3. Discuss the patient’s previously expressed end-of-life wishes or
documentation thereof.
4. Discuss remaining treatment options or lack thereof for the life-limiting illness.
5. Discuss what an acceptable quality of life would be for a patient.
48 Neurocognitive Disorders
progressive conditions that impact length and quality of life. A good palliative care
program is meant to improve symptom management, enhance end-of-life education and
shared decision-making, provide additional emotional and psychological support for
patients and families, better coordinate care, provide a holistic approach, and ease the
transition to end-of-life care when appropriate. See Figure 10.1.
Hospice care is appropriate for patients that are entering the terminal stage of illness
and is provided through a licensed hospice agency. The hospice care model focuses on
improving comfort and quality of life. It is a holistic approach that encompasses physical,
emotional, social, and spiritual support. Hospice care can be provided in almost any care
setting including long-term care, assisted living, or private homes. Hospices provide the
essential supplies and equipment that are necessary for end-of-life care such as hospital
beds, specialty mattresses, Hoyer lifts, wheelchairs, and wound care supplies, among
many others. It provides regular nursing assessments and care by nurses specifically
trained in end-of-life care. Services through hospice are covered through a patient’s
Medicare or Medicaid benefits or through a private insurance plan. Some hospice
agencies provide free care for those who have no funding source.
Hospice services are not appropriate for all patients and they must have one or more
hospice-eligible diagnoses (see Box 10.3). Hospice patients and their families should also
be “hospice minded”. This refers to a mindset or perspective that aligns with the
principles and values of hospice care, with patient-centered care focusing on comfort
and symptom management above likely futile disease-focused management. A useful
decision tree for determining hospice eligibility is presented in Figure 10.2.
Discussions of code status are delicate but important to have with patients and their
surrogate decision-makers. Code status refers to a patient’s wishes to undergo “heroic
measures” such as CPR, intubation with mechanical ventilation, and advanced life
support in the event of cardiopulmonary collapse. Patients and caregivers should be
informed that such heroic measures are much less likely to be successful in cases of
terminal illness and may be contrary to efforts to comfort the dying and ease the
transition from life to death. These conversations may be ongoing with patients and
families and can be seen as continuing opportunities to educate them on the
dying process.
“I Saw the Heavenly Gates” 49
The interventions of artificial hydration and nutrition in those with late-stage MNCDs
can be important to discuss with patients and their families. Primary caregivers are often
concerned with their loved one uncomfortably “starving to death” at the end of life.
In reality, these conversations can be opportunities to educate families that artificial
nutrition and hydration are not in fact adding comfort to the patient in the last days or
weeks of life. The absence of food and fluid intake results in ketosis and a release of opioids
in the brain, which may produce a sense of comfort or even euphoria [1]. Additionally, the
physiologic stress of processing nutrition will eventually be more than the multiple failing
organs can handle during the dying process, thereby hastening the end result.
In summary, physicians and other healthcare providers involved in direct patient care
of older adults should have a baseline knowledge of end-of-life care topics. Alternatively,
many hospice and palliative care physicians and APPs are easily available for consult-
ation and guidance on these topics. Providers should strive to provide excellent care to
patients during both life and the dying process.
50 Neurocognitive Disorders
Take-Home Points
Primary healthcare providers and mental health professionals are likely to be engaged with family
or care staff with questions regarding end-of-life care issues in those in the terminal stages of
MNCD. Those who provide care in the long-term care setting should be familiar with the issues
regarding end-of-life care in MNCD.
Hospice care is appropriate for patients who are entering the terminal stage of illness and is
provided through a licensed hospice agency. The hospice care model focuses on improving
comfort and quality of life. It is a holistic approach that encompasses physical, emotional, social,
and spiritual support. Hospice care can be provided in almost any care setting.
Palliative care is an approach to medical care that focuses on relieving pain, other symptoms, and
the physical, emotional, and psychosocial stress that individuals with serious illnesses may experi-
ence. The primary goal of palliative care is to improve the quality of life for patients and their
families, particularly when dealing with complex, chronic, or terminal medical conditions.
Discussions of code status are delicate but important to have with patients and the surrogate
decision-makers. Patients and caregivers should be informed that heroic measures are much less
likely to be successful in cases of terminal illness and may be contrary to efforts to comfort the
dying and ease the transition from life to death.
Mo, L., Geng, Y., Chang, Y. K., Philip, J., Parast, L., Tolpadi, A. A., Teno, J., Elliott,
Collins, A., & Hui, D. (2021). Referral M. N., & Price, R. A. (2022). Variation in
criteria to specialist palliative care for hospice experiences by care setting for
patients with dementia: A systematic patients with dementia. Journal of the
review. Journal of the American Geriatrics American Medical Directors Association,
Society, 69 (6), 1659. https://doi.org/10 23 (9), 1480. https://doi.org/10.1016/j
.1111/jgs.17070 .jamda.2022.03.010
Section 1 Neurocognitive Disorders
“Leave Me Alone”
Case
Mrs. L was an 82-year-old married woman living in a nursing home who had chronic
pain, MNCD-mixed-vascular and Alzheimer’s type, and chronic major depressive dis-
order. She had been doing well taking antidepressants (mirtazapine 15 mg at bedtime and
duloxetine 60 mg daily) and using a fentanyl pain patch (25 mcg/hour every three days) for
more than six months but started showing drowsiness and unsteadiness over the course of
two days.
Staff reported that Mrs. L had been shouting at various staff members, “Leave me
alone. I don’t feel like getting up.” The staff called her provider to request as-needed
lorazepam to calm her or to reduce her “psych medications” due to drowsiness. The
provider recommended an urgent same-day assessment as this behavior was new, of
sudden onset, and uncharacteristic. Mrs. L was calm and pleasant during the visit and the
provider asked Mr. L if he was aware of Mrs. L’s drowsiness and aggressive behavior.
Mr. L replied that he had figured out why Mrs. L’s condition had changed. He had
checked Mrs. L’s pain patch and found that the staff had forgotten to remove the
previous one when they put on a new patch two days before.
Teaching Points
Although psychiatric medications may sometimes be responsible for sedation and
unsteady gait, we recommend a thorough assessment to identify various other potential
causes of an acute change in behavior. The healthcare provider should always keep the
possibility of medication error in the differential diagnosis, especially if the patient has
been stable for some time. Additionally, an informed and astute caregiver (the husband
in this case) can often help identify the cause of an acute change in behavior quickly.
The proper assessment and treatment of acute and chronic pain in those with MNCD
has a strong influence on mental health. One study found the prevalence of pain from
osteoarthritis alone in those over 85 years old to be 72% [1]. Another study estimated
that between 40% and 80% of people living with dementia in long-term care homes
experience significant acute or chronic pain [2].
Common sources of pain in older adults in the long-term care setting include osteo-
arthritis/degenerative joint disease, peripheral neuropathy, peripheral vascular disease,
osteoporosis with compression fractures, cancer, and pressure wounds. Older adults may
present additional challenges to the medical provider when evaluating chronic pain. Some
patients may tend toward stoicism and be hesitant to discuss pain issues with their
providers. Others may fear the treatment for chronic pain. Some patients are very afraid
of addiction, even in the absence of any personal history of substance use or dependence.
52
“Leave Me Alone” 53
Table 11.1 Options for pharmacologic pain management in long-term care settings [9,10].
Take-Home Points
The implications of unrecognized pain in those with MNCD are far-reaching. Untreated pain can
lead to the destabilization of psychiatric conditions such as anxiety, depression, and agitation.
Those with cognitive impairments are more likely to have longer waits for the evaluation of pain
and receive less than adequate analgesia.
Always keep the possibility of medication error in the differential diagnosis for apparent changes in
mental status.
56 Neurocognitive Disorders
Those with MNCD with inadequately controlled pain may present with atypical symptoms when
compared to those without MNCD.
As-needed pain medications given to those with MNCD in the long-term care setting may depend
too heavily on the judgment of those inadequately trained to assess pain. Regularly scheduled pain
medication may be a more appropriate option.
Consider both pharmacologic and nonpharmacologic interventions when formulating a treatment
plan for chronic pain in older adults. Opiates may have the best benefit/risk ratio for the treatment
of chronic pain in some older adults.
Mr. S. was an 80-year-old white male with a history of atrial fibrillation, hypertension,
Type 2 diabetes, and major depressive disorder (MDD). He was originally admitted to
long-term care as he lived alone and was no longer able to look after himself. The
consultation with geriatric psychiatry was initially requested as Mr. S had been recently
noted to be calling his daughter and making statements of fear regarding his feelings that
someone was out to harm him or that people were intentionally ignoring him.
He reported to the consulting psychiatrist that he was having intermittent feelings of
sadness and was feeling guilty with regard to choices he had made in his younger days.
He denied any suicidal or homicidal thoughts, paranoia/delusions, or hallucinations on
the day of initial psychiatric evaluation. At the time of the initial evaluation he was on
escitalopram 5 mg daily, which was increased to 10 mg daily with thoughts that the
patient’s MDD was only in partial remission and would respond to a higher dose of SSRI.
As time went on Mr. S mentioned several times that he was lonely and felt little ability to
connect with others. His one living family member was his daughter, who lived six or
more hours away and was only able to see Mr. S once a month or so.
As time went on the increase in SSRI dose, along with weekly psychotherapy, helped
the patient’s mood symptoms improve. He began to participate in more social activities
and started to foster some friendships. After about eight months psychiatry was asked to
reevaluate Mr. S as he was again having increasing symptoms of depression. In recent
months he had been switched by his primary provider from escitalopram 10 mg daily to
sertraline 50 mg daily and mirtazapine 15 mg qhs.
His depression was exacerbated by an abrupt worsening of vision in his right eye.
He was already blind in his left eye and was found to have a retinal bleed that made him
legally blind. He felt more isolated as his blindness left him much less able to participate
in social or recreational activities in the facility. He was also less able to participate in his
own care and had become almost totally dependent on caregivers.
Anxiety had become more prominent. His sertraline was increased to 100 mg daily,
mirtazapine continued, and buspirone was titrated up to 10 mg tid. Despite medication
adjustment, Mr. S continued to become more despondent and was noted to perseverate
on his loss of sight and increasing dependence on others. He began making regular
passive suicidal statements. His sertraline was stepped up further to 200 mg daily.
During a follow-up visit it was discovered that the patient had a love of classical
music. The clinician and patient would spend 30 minutes during visits listening to and
discussing their favorite classical music pieces. These interactions seemed to open the
patient up to further brief motivational interviewing with a focus on adapting to his new
disabilities in positive ways to help his mood and overall outlook. On the next couple of
57
58 Mood Disorders
Teaching Points
This is a complex case for several reasons. It was felt that the patient never fully adjusted
to his move from his home in the community to the nursing home. His limited family/
social support was his daughter, who lived hours away. Mr. S was still cognitively intact
and being surrounded by others he saw as cognitively worse off than himself was a
source of stress. Despite being in a facility full of friendly residents and staff he had
difficulty mixing into the community. The unfortunate development of near-total
blindness from a retinal bleed further exacerbated the patient’s depression and isolation.
Several appropriate medication interventions were trialed over the course of months
with Mr. S. He was started on escitalopram and the dose was increased to the maximum
dose recommended in geriatric patients (10 mg). When this proved ineffective, he was
switched by his primary team to sertraline. The sertraline was increased to the maximum
dose and augmentation was later started with buspirone and then later aripiprazole. This
was a clinically sound progression of medication management of MDD with anxiety in a
geriatric patient. Consideration was then given to ECT.
“I Am in Hell and I Will Die in Hell” 59
3. Schizophrenia with:
a. Severe psychosis resistant to antipsychotics.
b. Severe mood symptoms or severe agitation resistant to medications.
c. Catatonia.
Take-Home Points
Reasonable strategies to address TRD in older adults include adding an antidepressant in another
class or adding one or more of many available augmentation agents.
Electroconvulsive therapy is thought by many to be underutilized in those with TRD. Treatment
responses to ECT can be brisk and dramatic.
Mr. S. was an 80-year-old white male with a history of atrial fibrillation, hypertension,
Type 2 diabetes, and major depressive disorder (MDD). He was originally admitted to
long-term care as he lived alone and was no longer able to look after himself. The
consultation with geriatric psychiatry was initially requested as Mr. S had been recently
noted to be calling his daughter and making statements of fear regarding his feelings that
someone was out to harm him or that people were intentionally ignoring him.
He reported to the consulting psychiatrist that he was having intermittent feelings of
sadness and was feeling guilty with regard to choices he had made in his younger days.
He denied any suicidal or homicidal thoughts, paranoia/delusions, or hallucinations on
the day of initial psychiatric evaluation. At the time of the initial evaluation he was on
escitalopram 5 mg daily, which was increased to 10 mg daily with thoughts that the
patient’s MDD was only in partial remission and would respond to a higher dose of SSRI.
As time went on Mr. S mentioned several times that he was lonely and felt little ability to
connect with others. His one living family member was his daughter, who lived six or
more hours away and was only able to see Mr. S once a month or so.
As time went on the increase in SSRI dose, along with weekly psychotherapy, helped
the patient’s mood symptoms improve. He began to participate in more social activities
and started to foster some friendships. After about eight months psychiatry was asked to
reevaluate Mr. S as he was again having increasing symptoms of depression. In recent
months he had been switched by his primary provider from escitalopram 10 mg daily to
sertraline 50 mg daily and mirtazapine 15 mg qhs.
His depression was exacerbated by an abrupt worsening of vision in his right eye.
He was already blind in his left eye and was found to have a retinal bleed that made him
legally blind. He felt more isolated as his blindness left him much less able to participate
in social or recreational activities in the facility. He was also less able to participate in his
own care and had become almost totally dependent on caregivers.
Anxiety had become more prominent. His sertraline was increased to 100 mg daily,
mirtazapine continued, and buspirone was titrated up to 10 mg tid. Despite medication
adjustment, Mr. S continued to become more despondent and was noted to perseverate
on his loss of sight and increasing dependence on others. He began making regular
passive suicidal statements. His sertraline was stepped up further to 200 mg daily.
During a follow-up visit it was discovered that the patient had a love of classical
music. The clinician and patient would spend 30 minutes during visits listening to and
discussing their favorite classical music pieces. These interactions seemed to open the
patient up to further brief motivational interviewing with a focus on adapting to his new
disabilities in positive ways to help his mood and overall outlook. On the next couple of
57
58 Mood Disorders
Teaching Points
This is a complex case for several reasons. It was felt that the patient never fully adjusted
to his move from his home in the community to the nursing home. His limited family/
social support was his daughter, who lived hours away. Mr. S was still cognitively intact
and being surrounded by others he saw as cognitively worse off than himself was a
source of stress. Despite being in a facility full of friendly residents and staff he had
difficulty mixing into the community. The unfortunate development of near-total
blindness from a retinal bleed further exacerbated the patient’s depression and isolation.
Several appropriate medication interventions were trialed over the course of months
with Mr. S. He was started on escitalopram and the dose was increased to the maximum
dose recommended in geriatric patients (10 mg). When this proved ineffective, he was
switched by his primary team to sertraline. The sertraline was increased to the maximum
dose and augmentation was later started with buspirone and then later aripiprazole. This
was a clinically sound progression of medication management of MDD with anxiety in a
geriatric patient. Consideration was then given to ECT.
“I Am in Hell and I Will Die in Hell” 59
3. Schizophrenia with:
a. Severe psychosis resistant to antipsychotics.
b. Severe mood symptoms or severe agitation resistant to medications.
c. Catatonia.
Take-Home Points
Reasonable strategies to address TRD in older adults include adding an antidepressant in another
class or adding one or more of many available augmentation agents.
Electroconvulsive therapy is thought by many to be underutilized in those with TRD. Treatment
responses to ECT can be brisk and dramatic.
13 Antipsychotic-Related Movement
Disorders
Ms. R was a 77-year-old female who had been a resident in long-term care for almost two
years. Prior to living in long-term care she lived in a local senior housing building and
retained her independence there with the help of her daughter. Her daughter would visit
daily and make sure her mother had all her basic needs met and was receiving her
medications as prescribed, but was forced to move an hour away because of work.
Ms. R had a lifelong history of bipolar disorder. She was diagnosed in her early 30s, at
which time she was hospitalized for two to three weeks following her first manic episode.
She was on and off her medications frequently and had recurrent hospitalizations related
to this throughout her adult life. She became more stable in her 50s when she qualified
for federal disability income and her life became less stressful and more routine.
Ms. R had a happy childhood. She had a loving home with two parents and two
siblings. Her father was a Methodist minister and her mother stayed at home, taking care
of the children and home. She did well in school and went to college for two years where
she studied office management. She had been a rather accomplished pianist as well. She
married her high school sweetheart and had a happy marriage. She was employed to run
a dental office for some years prior to showing signs of extreme stress from both her
work and family obligations. She began using over-the-counter weight loss pills to “help
me keep up”. She then began having difficulty keeping up with her work responsibilities
and was eventually hospitalized for a mental breakdown and diagnosed as being “manic
depressive”.
As Ms. R approached her mid-60s, again she began having more frequent hospital-
izations for her bipolar disorder. She was hospitalized on average once every year or two.
Her medications were adjusted often as her lithium monotherapy became less and less
effective at mood stabilization. She would be stable on a particular combination of
medications for an average of two to three years before major medication adjustments
would be necessary
At the age of 75, Ms. R moved into a Medicaid-funded bed at a long-term care facility
in her small town after one of her psychiatric hospitalizations. This was at the urging of
her daughter who was no longer able to provide adequate care oversight in her home due
to work obligations. Ms. R settled in surprisingly well at the facility. She was described as
sweet and friendly by all the staff at the facility, and she had many friends.
Two years after moving into long-term care, Ms. R started behaving erratically and
becoming quite irritable. Her sleep patterns became unusual and she began having
conversations with herself, or at times appeared to be speaking to the wall, according
to staff. Complicating the situation was the fact that her facility psychiatrist had retired
and there were no other psychiatrists that serviced the small town where she lived. She
62
“My Mom Looks Like a Zombie!” 63
was transferred to the city and was hospitalized for two weeks for an acute manic
episode. She was discharged on divalproex 750 mg twice daily and aripiprazole 15 mg
qhs, along with prn lorazepam 1 mg q6 hours for anxiety.
Ms. R’s daughter came to visit her at the facility and expressed her shock at her
mother’s appearance. “My mom looks like a zombie!” At the first physician visit post-
discharge, she appeared quite different to before her hospitalization. Ms. R was now
dependent on a wheelchair for mobility when she had been using a walker before her
hospitalization. Her face had a drawn and frozen appearance, her extremities were stiff,
and she had a subtle slow tremor noted in her left hand. Ms. R was also much less
interactive, and her cognition had slowed. She had a hard time feeding herself and
needed help from staff to eat her meals.
The long-term care provider examined Ms. R and reviewed all of her recent hospital
documents. She had a head CT with her recent hospitalization that was negative for any
acute findings and showed mild age-appropriate changes. Ms. R was checked for a UTI
and had her labs checked again, but they did not reveal any acute metabolic issues. Her
exam was consistent with Parkinsonism and her long-term care provider thought it likely
that these symptoms were induced by the recent addition of aripiprazole as a mood
stabilizer during her hospitalization. Her dose of aripiprazole therefore was tapered over
two weeks to 5 mg nightly. This improved the Parkinson’s features, but Ms. R again
began showing signs of impending mania with increased agitation and decreased need
for sleep. After consulting with the long-term care provider, Ms. R’s daughter requested
something be done for the mania short of another hospitalization. The long-term care
provider started Ms. R on quetiapine and titrated the dose to 200 mg twice daily. This
seemed to avert another manic episode, but Ms. R continued to demonstrate
Parkinsonian characteristics. The patient and her daughter were disappointed at the side
effects of her medication but were very pleased with the good control of her
bipolar disorder.
Teaching Points
Antipsychotic medications are part of the pharmacologic treatment of many psychiatric
disorders, including schizophrenia, schizoaffective disorder, bipolar disorder, MDD, and
delusional disorder, among others. Much of the management of these conditions is
through a mental health specialist such as a psychiatrist or psychiatric nurse practitioner.
But many places in the US are underserved by mental health specialists and long-term
care providers may be required to provide psychiatric care to their patients.
Some of the more difficult challenges that antipsychotic providers may face are side
effects involving drug-induced movement disorders. These are collectively referred to as
extrapyramidal side effects (EPS) and are due to the dopamine blocking mechanism of
the antipsychotic class of medications.
There are four well-known extrapyramidal symptoms that the provider should be
familiar with (see Box 13.1). Three of these presentations can be seen within days and
weeks after initiating antipsychotic treatment and include acute dystonia, akathisia, and
Parkinsonism. Tardive akathisia and tardive dyskinesia (TD) are considered manifest-
ations of chronic antipsychotic use. Extrapyramidal side effects are more likely to occur
with older first-generation antipsychotic medications and are seen less commonly now
that the use of second-generation antipsychotics far surpasses older first-generation
64 Mood Disorders
antipsychotics. The majority of newly-diagnosed EPS are now in patients who are on
atypical second-generation antipsychotics.
Acute dystonia is a relatively uncommon occurrence and usually happens early in the
treatment with an antipsychotic. Most of the acute dystonic reactions involve the muscles
of the face and neck. Box 13.2 lists some common manifestations of acute dystonia
related to antipsychotic use.
The treatment of acute dystonic reactions is the swift administration of an antihista-
mine such as diphenhydramine or benztropine. In the emergency room, these are usually
given intramuscularly or intravenously and often produce a swift and dramatic relief of
symptoms. Strategies to handle the need for antipsychotics in patients with a history of
acute dystonic reactions include prophylactic or ongoing use of an antihistamine medi-
cation such as benztropine, using the lowest dose necessary to control the side effects.
Akathisia is a distressing and uncomfortable movement disorder in which patients
have a feeling of restlessness and an objective increase in motor activity. People with
akathisia often experience an inner sense of restlessness, a strong urge to move, and an
“My Mom Looks Like a Zombie!” 65
inability to sit still. These sensations can be quite distressing and may lead to significant
discomfort and agitation. Patients can demonstrate akathisia in several ways. Leg rest-
lessness is common and can be seen as continuous tapping of the feet, pacing, or
constantly shifting weight from one leg to the other while standing. It can also be seen
when sitting as frequent shifting in the seat or rocking back and forth. Patients with
akathisia may also demonstrate an almost complete inability to stand or sit still.
Restlessness can also lead to increased anxiety and agitation.
Several interventions can help manage akathisia. Lowering the dose of the antipsy-
chotic can be helpful if this is possible and switching the antipsychotic to an alternative
agent may be useful. If these interventions are not successful several medication options
may help to lessen the akathisia. These include beta-blockers (propranolol) and
benzodiazepines. Beta-blockers such as propranolol may be effective at lower doses
and tolerated by older adults. Benzodiazepines may be effective in ameliorating symp-
toms of akathisia in younger long-term care residents but should be avoided if possible
in older adults. Trazodone has shown promise in the treatment of akathisia and is well
tolerated in older adults [1]. Low-dose mirtazapine (7.5 mg daily) has also shown
promise as a potential treatment for akathisia and is generally well tolerated in older
adults.
Parkinsonism related to antipsychotic use clinically looks quite similar to the symp-
toms of Parkinson’s disease with features such as tremors, muscle rigidity, bradykinesia,
and postural instability. It can be seen with the use of both first- and second-generation
antipsychotics and can develop acutely or gradually. These are relatively common with
antipsychotic medications but are much more likely with potent first-generation anti-
psychotics such as haloperidol. It is estimated that over 50% of patients over 60 on long-
term therapy with antipsychotics will experience medication-related Parkinsonism [2].
Drug-induced Parkinsonism is generally treated by stopping the offending antipsychotic
medication. If an antipsychotic is required one should be utilized that is known to have
less risk of drug-induced Parkinsonism. These agents will have less affinity for dopamine
d2 receptors and include quetiapine, clozapine, olanzapine, and brexpiprazole [3].
Parkinsonism usually dissipates within weeks to months after discontinuing the
offending agent, but 10–50% of patients may retain Parkinsonian features even after
the medication is discontinued. Some of the Parkinsonism seen in older patients
on antipsychotics may also suggest a possible unmasking of a previously preclinical
Parkinson’s disease.
Tardive dyskinesia is considered a late-occurring side effect of antipsychotic medica-
tions. It most frequently involves involuntary repetitive movements of the facial muscles
but can also affect the neck, hands, limbs, and even walking. It is one of the most feared
complications of chronic antipsychotic use as it can dramatically affect a patient’s quality
of life and lead to social withdrawal and increased depression and anxiety. Older adults
have been shown to be at higher risk of TD than younger patients. This is true even when
lower doses and shorter courses of antipsychotics are used [4]. Tardive dyskinesia
symptoms include grimacing, lip smacking, tongue movements, uncontrolled chewing
or teeth grinding, blinking, and arrhythmic head or limb movements. It is hard to treat
and often irreversible. Therapeutic strategies to address TD may include changing to
another antipsychotic medication less inclined to be associated with TD, adjusting the
dose of the implicated medication, or utilizing medications indicated to treat TD.
Box 13.3 reiterates strategies to deal with TD.
66 Mood Disorders
Take-Home Points
Teaching Points
The primary care provider is often the first clinician to recognize the high burden of life-
prolonging treatment for a resident who has limited life expectancy and high medical
and neurocognitive comorbidity. It is important to explain to residents, their families,
and staff about the significant burdens imposed by hospitalization, steroids, and antibi-
otics on residents who have advanced MNCD. The value of palliative care goals is
ensuring that the last weeks and months of life are peaceful, dignified, and without the
burden of excessive treatment. The management of delirium also offers an opportunity
for the primary provider to provide case-based education that is tailored to the staff’s
educational needs (in this case, poor recognition of the resident’s limited life expectancy,
limited understanding of the high burdens of life-prolonging care on many residents,
and lack of recognition of the importance of inquiring into Mrs. S’s values and wishes
regarding palliative care).
There is much crossover in the fields of palliative care and psychiatry. Palliative care
refers to specialized medical care provided to individuals with serious illnesses or
conditions that are not curable. The goal of palliative care is to improve the quality of
life for patients by alleviating symptoms, managing pain, and addressing the physical,
emotional, social, and spiritual needs associated with their condition. Common psychi-
atric syndromes that are encountered in palliative care patients include anxiety with
agitation, depression, delirium, insomnia, and others. The approach to psychiatric care
in the palliative care setting can be somewhat different as goals of care often focus more
on patient comfort than on life-prolonging interventions.
Causes of anxiety in palliative care patients can be categorized as primarily psychi-
atric, organic, iatrogenic, and existential. Psychiatric causes of anxiety include poorly
controlled underlying anxiety or mood disorder or PTSD, for example. Organic causes of
anxiety include underlying physical processes such as hypoxia, pain, or sepsis. Examples
of iatrogenic causes of increased anxiety would include supratherapeutic doses of thyroid
replacement or the use of psychiatric medications that have a potential to be over-
stimulating. Existential causes of anxiety include concerns about increasing dependency,
disfigurement, the meaning of one’s life, leaving one’s loved ones, and concerns about
what happens after death. Psychosocial stressors can also be contributors to increased
anxiety (see Box 14.1).
Clinical depression is also relatively common at the end of life. The prevalence of
MDD at the end of life for cancer patients is estimated to be between 5–20% [1]. The
contributors to depression at the end of life include: (1) Grief and loss: The awareness of
impending death and the experience of significant losses, such as declining health,
independence, or the loss of relationships, can trigger feelings of grief and sadness.
This grief can manifest as depressive symptoms. (2) Physical discomfort: Pain, fatigue,
and other physical symptoms associated with advanced illness can contribute to a decline
in overall well-being, affecting mood and potentially leading to depressive symptoms. (3)
Social isolation: The end-of-life phase may involve changes in social support networks, as
individuals may become more confined to home or require specialized care. Social
isolation or a decreased ability to engage in meaningful social interactions can contribute
to feelings of loneliness and depression. (4) Existential distress: Reflecting on mortality
and the meaning of life can evoke existential questions and concerns. Existential distress,
including feelings of hopelessness, regret, or a loss of purpose, can contribute to
70 Mood Disorders
4. Psychosocial stressors:
a. dysfunctional family dynamics.
b. unmet financial needs.
c. concerns about advancing caregiving needs.
d. social isolation at the end of life.
e. legal matters at the end of life.
f. caregiver burden.
Box 14.2 Considerations in managing mood symptoms for patients nearing end of life
1. Have a high index of suspicion for depression or anxiety at the end of life.
2. Realize that depression and anxiety are not expected complications of end of life.
3. Early recognition and treatment of depression and anxiety can reduce significant
suffering in those at the end of life.
4. Make sure that physical symptoms at the end of life are adequately controlled. Anxiety
and depression can be directly related to poorly controlled symptoms such as pain
or dyspnea.
“My Soul Is in England” 71
Take-Home Points
The primary care provider is often the first clinician to recognize the high burden of life-prolonging
treatment for a resident who has limited life expectancy and high medical and
neurocognitive comorbidity.
Clinical depression is relatively common at the end of life. The prevalence of MDD at the end of life
for cancer patients is estimated to be up to 20%.
Palliative care refers to specialized medical care provided to individuals with serious illnesses or
conditions that are not curable. The goal of palliative care is to improve the quality of life for
patients by alleviating symptoms, managing pain, and addressing the physical, emotional, social,
and spiritual needs associated with their condition.
Choose psychotropic medications that have a quicker response time when possible. Examples
include stimulants for depressive symptoms and benzodiazepines for anxiety. The use of SSRIs,
SNRIs, and other more commonly prescribed medications for depression and anxiety may take up
to four to six weeks for clinical response.
“I Feel Terrible”
Case
Mrs. U, a 90-year-old resident in a nursing home who had MNCD due to Alzheimer’s
disease, had been experiencing severe depressive symptoms for four months. Symptoms
included tearfulness, lack of interest in activities, severe anxiety, insomnia, lack of
appetite, weight loss, and irritability. A referral to a psychiatrist was made as
Mrs. U had not responded to two adequate trials of antidepressants. The psychiatrist
found that the patient had been tried on citalopram 20 mg daily for eight weeks and then
sertraline 50 mg for eight weeks before the referral. The patient’s primary care physician
was reluctant to use higher doses of antidepressants because of the patient’s advanced age
and MNCD.
In the week before evaluation by the psychiatrist, Mrs. U had developed lower
abdominal pain and was found to have a UTI so was put on antibiotics. At the time of
the interview, the psychiatrist found her to be severely depressed and tearful throughout
the interview, expressing statements indicating hopelessness and a wish that she was
dead. Mrs. U was also distraught because she had developed mild antibiotic-induced
diarrhea. She told the psychiatrist, “I feel terrible.” Her SLUMS score was 13. The
psychiatrist considered restarting one of the SSRIs but because of the risk of worsening
antibiotic-induced diarrhea, decided to avoid them and start mirtazapine instead.
Mrs. U was put on 7.5 mg of mirtazapine daily at bedtime and the dose was increased
after seven days to 15 mg daily at bedtime. She started sleeping better after two weeks of
treatment, the diarrhea resolved when the antibiotic course was over, the UTI resolved,
and the abdominal pain improved. However, her tearfulness, daytime agitation, and
hopelessness persisted. Hence, the dose of mirtazapine was further increased to 22.5 mg
daily at bedtime for seven days. Mrs U. was able to tolerate this dose without sedation or
other adverse effects.
An individualized, pleasant activity schedule was created and initiated for Mrs. U,
after consultation with the family and staff. The family and staff were counseled to try to
walk with her daily as she had been active all her life, and bright light therapy was started
(exposure to bright light for one hour every morning while Mrs. U was perusing her
favorite magazines). Mrs. U enjoyed hugs from the staff, and staff who were comfortable
were encouraged to hug her off and on throughout the day. Mrs. U enjoyed classical and
instrumental music, and listening to this music was added to her daily schedule. The
family hired a massage therapist to come twice a week. After eight weeks, her mood had
improved significantly, her anxiety decreased, her appetite had returned to near normal,
and her feelings of hopelessness had resolved.
73
74 Mood Disorders
Teaching Points
This case provides the opportunity to examine some common treatment challenges in
depression for older adults. One of the first dilemmas is often which antidepressant to
choose as initial treatment for newly-diagnosed MDD. Current guidelines still support
the use of an SSRI as the first choice for the treatment of depression in older adults.
Although no SSRI has been shown to be more efficacious than other SSRIs, sertraline is
the favorite choice of this author due to its safety, tolerability, effectiveness, and ability to
titrate the dose. The potential benefits and challenges of individual SSRIs are listed in
Table 15.1.
A second question is how to proceed in patients who do not have an initial full
and positive remission with a dose of sertraline that has reached a therapeutic
dose. A potential treatment algorithm for newly-diagnosed MDD is represented in
Box 15.1 [1].
Clinicians should keep in mind the old adage “start low, go slow, but go” with regard
to the prescribing of antidepressants in older patients. With older adults, it is a good idea
to start at half or quarter of the normal adult starting dose but to increase the dose to well
within the therapeutic range for a given antidepressant. This escalation to a therapeutic
Table 15.1 Potential positives and negatives to the available generic SSRIs.
Box 15.1 Treatment suggestion for new diagnosis depression in older adults
1. Start with an SSRI such as sertraline at a dose that is a half or quarter of the normal
adult starting dose (12.5 or 25 mg daily).
2. Institute patient-specific nonpharmacologic interventions such as psychotherapy,
recreation therapy, exercise, pet therapy, massage, increased family involvement, and
so on.
3. In two weeks increase the dose of sertraline to 50 mg daily.
4. Reassess every four weeks for response or remission of symptoms. If there is an
inadequate response, consider increasing the dose of sertraline by 25–50 mg/day to a
max dose of 200 mg daily.
5. Reassess in four weeks. If inadequate response, initiate a trial of aripiprazole 2 mg
daily [1].
6. Reassess in four weeks. If inadequate response, increase the dose of aripiprazole to
5 mg daily.
7. If there is an inadequate response, after four weeks the patient can be considered
treatment resistant. At this point, a consultation with a psychiatrist may be warranted.
If not available, consider proceeding to step 8.
8. Consider adding a third agent from a different antidepressant class such as an SDRI
(bupropion) or SNRI (venlafaxine, duloxetine, or desvenlafaxine). Start at half of the
normal adult starting dose.
9. Titrate the dose of the SDRI or SNRI to mid to upper therapeutic dose by small steps
every two to four weeks.
10. If the patient does not respond adequately to treatments at this point it may be
advisable to consult a geriatric psychiatrist.
dose should happen slowly over the course of one to two months. If an older patient does
not respond to a typical therapeutic dose the clinician should not feel overly cautious
about prescribing a dose at the upper limit of FDA-approved doses.
This case also highlights the nonpharmacologic interventions important in the
treatment of depression. Pharmacologic treatments have a higher likelihood of failing
in the absence of nonpharmacologic interventions.
Loneliness is an epidemic in the lives of older adults and can contribute greatly to the
development and persistence of mood and anxiety disorders. Loneliness can be due to
both inadequate quantity and quality of social relationships [2,3]. Insufficient quantity
and poor-quality relationships can be problematic even in a long-term care setting with a
relatively large number of residents and staff. Boredom can also significantly impact
mental health. The more successful long-term care facilities have active social, spiritual,
and recreational activity programs to enhance connections and give more meaning to the
life of seniors.
The healing effect of human touch is also highlighted in this case study. Studies have
shown that psychosocial interventions can be as effective or more effective than pharma-
cological therapies for the treatment of depression [4]. Animal therapy, cognitive
stimulation, massage therapy, and reminiscence therapy should be considered as options
for supportive interventions in those with depression in long-term care [5]. There can be
time, financial, and access issues to these services in long-term care facilities that may
make utilization of these interventions problematic.
76 Mood Disorders
Take-Home Points
Depression is prevalent in older adults in long-term care with an estimated prevalence of 11–45%.
Depression is associated with poor overall health outcomes and poor quality of life. Interventions
are often effective if these patients are identified, but treatment can be challenging and require
multiple adjustments in medications or treatment strategies.
Current guidelines still support the use of an SSRI as the first choice for the treatment of depression
in older adults.
Clinicians should keep in mind the old adage “start low, go slow, but go” with regard to the
prescribing of antidepressants in older patients. With older adults, it is a good idea to start at half or
quarter of the normal adult starting dose but to increase the dose to well within the therapeutic
range for a given antidepressant.
Clinical depression is relatively common at the end of life. The prevalence of MDD at the end of life
for cancer patients is estimated to be up to 20%.
“I Feel Terrible” 77
“I Don’t Care”
Case
Mr. B was an 82-year-old retired accountant living in a long-term care facility. He had
been experiencing a lack of interest in activities, decreased appetite, decreased energy,
and agitation during personal care. He had been expressing statements such as “I don’t
care” for three months. The symptoms started a few weeks after he was moved from an
AL home to a skilled nursing home due to advancing MNCD and recurrent falls. He also
had a recent history of GI bleeding due to peptic ulcer disease.
On clinical exam, the long-term care provider noted that Mr. B had a depressed
mood and poor eye contact. Major neurocognitive disorder due to Alzheimer’s disease
with MDD was diagnosed. The primary provider considered prescribing SSRIs but did
not due to Mr. B’s recent history of GI bleeding and the potential for an SSRI to increase
the risk of bleeding. Mirtazapine was considered but not prescribed because of its
propensity to cause weight gain and Mr. B had morbid obesity with related mobility
impairment. Bupropion was started at 75 mg daily in the morning and after seven days
increased to 75 mg in the morning and at 5 pm. An individualized, pleasant activity
schedule was created for Mr. B after a discussion with him, his family, and staff. The
family was encouraged to bring grandchildren to visit Mr. B as they always cheered him
up, and it was recommended that staff bring Mr. B’s former roommate to visit as they
had become close friends over the two years that they lived together in the AL home.
Over the next four weeks, Mr. B gradually started talking more and eating more. The
bupropion was changed to bupropion sustained-release preparation, given as 150 mg
once daily in the morning. After eight weeks Mr. B was attending activities and eating
better and the agitation during personal care had reduced dramatically.
Teaching Points
Major depressive disorder is a prevalent cause of disability worldwide and can cause
significant and enduring impairment in older adults. According to the Diagnostic and
Statistical Manual of Mental Disorders–5–TR (DSM–5–TR), MDD is characterized by a
duration of at least two weeks during which a patient experiences five or more of the
following symptoms nearly every day: depressed mood, lack of interest or pleasure
(anhedonia), changes in weight or appetite, sleep disturbances (insomnia or hypersom-
nia), psychomotor retardation, fatigue or loss of energy, feelings of worthlessness or
excessive guilt, difficulty concentrating or making decisions, and recurrent thoughts of
death, suicide, or a suicide attempt. The most prominent theory regarding the under-
lying mechanisms of MDD is the monoamine deficiency hypothesis, which proposes that
depression arises from the depletion of serotonin, norepinephrine, or dopamine in key
78
“I Don’t Care” 79
areas of the CNS. Among these neurotransmitters, serotonin has received considerable
attention in the study of depression’s pathophysiology. Research has shown that
reduced synthesis and effective concentrations of serotonin at the neural synapses in
key areas of the brain contributes to depressive symptoms in individuals vulnerable
to depression.
Antidepressants, when used appropriately and in combination with an individual-
ized psychosocial approach, can dramatically improve depressive symptoms and the
quality of life of residents who have MDD. The selection of antidepressants needs to
consider the patient’s medical problems as well as what side effects one wants or
wants to avoid for a particular patient. There is no compelling evidence that one
antidepressant works better than any other for the treatment of MDD in long-term
care populations. Selective serotonin reuptake inhibitors are probably the most
commonly selected first-line medications for the treatment of MDD in long-term
care residents. They function by impeding the reuptake of serotonin, thereby increas-
ing its availability in synapses. This mechanism of action within the CNS explains the
efficacy of SSRIs for depression but the effects are not limited to the CNS [1].
Serotonin transporters are present in tissues throughout the body. This fact helps
to explain why up to 80% of patients taking SSRIs will experience at least one side
effect. Some of the potential side effects include sexual dysfunction, anxiety, dizziness,
weight gain, GI problems, and headaches. Other adverse effects include prolongation
of the QT interval, coagulopathy, the potential for serotonin syndrome, and SSRI
discontinuation syndrome [2].
Increased bleeding risk associated with SSRI use has been reported, notably in the
upper GI tract. The existing literature on this subject reveals mixed findings: Some
studies indicate no elevation bleeding risk with SSRIs, others demonstrate an increased
risk of bleeding in the perioperative period when SSRIs are combined with NSAIDs [3].
The proof of risk of intracranial bleeding associated with SSRIs is lacking. The study by
Isokuortti et al. indicated no increased risk or intracranial bleeding in those on SSRIs
who sustained head trauma [4]. The study by Jensen et al. demonstrated insufficient data
to recommend restricting the use of SSRIs because of concern for increased risk of
intracranial bleeding complications [5]. The consensus currently seems to be that SSRIs
cause a true increase in GI bleeding complications but that these effects can be lessened
with certain precautions.
Serotonin–norepinephrine reuptake inhibitors have also been associated with a
potentially increased risk of bleeding, although the risk appears to be lower compared
to SSRIs. The potential for bleeding when using SNRIs is generally regarded as modest,
and most individuals still experience a favorable balance between the risks and benefits of
these medications [6].
The mechanism behind the impact of SSRIs on increased bleeding complications is
multifactorial. Inhibition of serotonin reuptake can influence platelet aggregation as
platelets also express the serotonin transporter. Selective serotonin reuptake inhibitors
can lead to reduced storage of serotonin in platelet-dense granules, potentially impacting
bleeding tendencies. Moreover, increased serotonin levels can stimulate gastric acid
secretion, thereby raising the risk of ulcers. When SSRIs are combined with NSAIDs
or antiplatelet agents, the risk of upper GI bleeding is significantly augmented. Examples
of commonly used antiplatelet agents include aspirin, clopidogrel, prasugrel, ticagrelor,
persantine, and cilostazol.
80 Mood Disorders
Despite the increased risk of bleeding with SSRIs and antiplatelet agents, there are
times when a patient may require an antiplatelet agent and it is thought that an SSRI may
still be the best antidepressant option. In these cases, the use of a proton pump inhibitor
(PPI) should be considered. Proton pump inhibitors have been shown to reduce the risk
of GI bleeding in those that have increased risk such as in those patients who require
NSAIDs or antiplatelet agents and SSRIs [2].
Some studies have shown an estimated 30–70% increased risk of bleeding when the
vitamin K antagonist warfarin is used along with SSRIs in hospitalized patients [7]. It is
hard to know if these findings can be generalized to a non-hospitalized patient popula-
tion as well. The utilization of PPIs may also be beneficial in these cases to help mitigate
the risk of combining this blood thinner with SSRIs.
Since 2010 the use of direct oral anticoagulants (DOACs) has increased. Examples of
these medications include apixaban (Eliquis), dabigatran (Pradaxa), and rivaroxaban
(Xarelto). Their safety and efficacy make them more popular choices for those who need
anticoagulation. Before the arrival of DOACs, warfarin was the only choice for oral
anticoagulation. Studies have shown an increase in bleeding risk when DOACs are
combined with SSRIs and for this reason patients requiring this treatment should be
considered for concomitant PPI use [8].
There are strategies to mitigate the increased bleeding risk that can be associated with
SSRIs and SNRIs (Box 16.1).
It should be mentioned that the pathophysiology of depression is still in active
research with alternative therapeutic targets having been identified in recent years.
Ketamine and esketamine have multiple pharmacodynamic targets but the major target
is the NMDA receptor, for which they act as an antagonist. By doing so, ketamine
modulates the release and function of various neurotransmitters, including glutamate, in
several brain regions. This modulation of glutamate transmission is thought to play a
crucial role in the antidepressant effects of ketamine. Another important downstream
effect of ketamine and esketamine is the rapid and transient increase in the release of
brain-derived neurotrophic factor (BDNF), a protein that promotes the growth, survival,
and function of neurons. Its deficiency has been associated with depression. The increase
in BDNF levels is believed to promote synaptic plasticity and the formation of new
connections between brain cells, which may contribute to the antidepressant effects of
ketamine. The roles of ketamine and esketamine are still being elucidated for the
management of TRD [9,10].
Box 16.1 Strategies to mitigate the increased risk of bleeding seen with SSRIs
and SNRIs
1. Antiplatelet agents and NSAIDs strongly increase the risk of upper GI bleeding with
SSRIs and SNRIs. Consider if a patient truly needs to be on an antiplatelet agent.
2. If a patient needs to be on an antiplatelet agent or NSAID and an SSRI or SNRI, consider
adding a PPI. This can help to decrease the risk of upper GI bleeding with SSRIs.
3. Consider an alternative antidepressant to an SSRI if the antiplatelet agent is necessary
and the patient has an increased risk of upper GI bleeding. Alternative agents to be
considered include bupropion, trazodone, and mirtazapine.
“I Don’t Care” 81
Take-Home Points
implementation. The American Journal of Jensen, M. P., Ziff, O. J., Banerjee, G., Ambler,
Psychiatry, 178 (5), 383. https://doi.org/10 G., & Werring, D. J. (2019). The impact of
.1176/appi.ajp.2020.20081251 selective serotonin reuptake inhibitors on
the risk of intracranial hemorrhage:
9. Edinoff, A. N., Akuly, H. A., Hanna,
A systematic review and meta-analysis.
T. A., Ochoa, C. O., Patti, S. J., Ghaffar,
European Stroke Journal, 4 (2), 144–152.
Y. A., Kaye, A. D., Viswanath, O., Urits,
I., Boyer, A. G., Cornett, E. M., & Kaye, Laporte, S., Chapelle, C., Caillet, P., Beyens,
A. M. (2021). Selective serotonin reuptake M. N., Bellet, F., Delavenne, X., &
inhibitors and adverse effects: A narrative Bertoletti, L. (2017). Bleeding risk under
review. Neurology International, 13 (3), selective serotonin reuptake inhibitor
387–401. https://doi.org/10.3390/ (SSRI) antidepressants: A meta-analysis of
neurolint13030038 observational studies. Pharmacological
Research, 118, 19–32.
10. Andrade, C., & Sharma, E. (2016).
Serotonin reuptake inhibitors and risk of Lee, T., Park, Y., Kim, S., You, H., Kang, J., &
abnormal bleeding. Psychiatric Clinics, Jung, Y. (2020). Concomitant use of
39 (3), 413–426. NSAIDs or SSRIs with NOACs requires
monitoring for bleeding. Yonsei Medical
Journal, 61 (9), 741–749. https://doi.org/10
Further Reading .3349/ymj.2020.61.9.741
Andrade, C., & Sharma, E. (2016). Serotonin McIntyre, R. S., Carvalho, I. P., Lui, L. M.,
reuptake inhibitors and risk of abnormal Majeed, A., Masand, P. S., Gill, H., &
bleeding. Psychiatric Clinics of North Rosenblat, J. D. (2020). The effect of
America, 39 (3), 413–426. doi: 10.1016/ intravenous, intranasal, and oral ketamine
j.psc.2016.04.010. Epub 2016 Jun 28. PMID: in mood disorders: A meta-analysis. Journal
27514297. of Affective Disorders, 276, 576–584.
Auerbach, A. D., Vittinghoff, E., Maselli, J., Quinn, G. R., Hellkamp, A. S., Hankey, G. J.,
Pekow, P. S., Young, J. Q., & Lindenauer, Becker, R. C., Berkowitz, S. D.,
P. K. (2013). Perioperative use of selective Breithardt, G., & Singer, D. E. (2018).
serotonin reuptake inhibitors and risks for Selective serotonin reuptake inhibitors and
adverse outcomes of surgery. JAMA bleeding risk in anticoagulated patients
Internal Medicine, 173 (12), 1075–1081. with atrial fibrillation: An analysis from the
doi: 10.1001/jamainternmed.2013.714. ROCKET AF trial. Journal of the American
PMID: 23699725; PMCID: PMC3867199. Heart Association, 7 (15), e008755.
Section 2 Mood Disorders
17 Depression/Psychogenomic Testing
Mr. G was a 79-year-old resident who had suffered from a stroke causing left hemipar-
esis. Staff requested that his long-term care provider evaluate Mr. G because of frequent
episodes of irritation, yelling at staff, making sexually inappropriate comments, and
attempting to touch staff inappropriately during personal care. These symptoms were
present for three months. Mr. G’s wife (and legal surrogate decision-maker) had refused
antidepressant treatment for Mr. G in the past because he had become “wild” after
initiation of an antidepressant (paroxetine) five years earlier after his first stroke.
Following a mental status exam, Mr. G showed evidence of executive dysfunction and
depressed mood. He enjoyed socialization and conversation with his provider and
seemed to crave social interaction and personal attention. Mr. G told the provider,
“Yes, I feel very lonely.” A diagnosis of poststroke depression and impulse control
disorder due to frontal lobe dysfunction was made. He admitted to feeling sad and
hopeless at times. He also expressed that he felt irritable.
The primary provider reviewed previous records, which indicated that Mr. G was
treated with 20 mg of paroxetine for depression five years earlier. The provider reviewed
the possible adverse effects of antidepressants with Mrs. G and the potential benefits of a
second trial with an antidepressant at a low dose. The nursing staff at the long-term care
facility encouraged Mrs. G to inquire about the use of pharmacogenetic testing “to see
which antidepressant is best”. The healthcare provider spoke with Mrs. G and encour-
aged her to proceed with a trial of one more antidepressant that was likely to be better
tolerated in older patients. The provider assured Mrs. G that Mr. G would be started on a
very low dose of medication, closely monitored for any adverse effects, and that the
antidepressant would promptly be discontinued if the adverse effect was moderate to
severe. Mrs. G agreed to this plan.
A decision was made to start Mr. G on sertraline 12.5 mg daily each morning, to be
increased to 25 mg daily in the morning after seven days. Mr. G developed transient mild
nausea and anxiety for three days, which resolved spontaneously. After treatment for
four weeks, the staff noticed that Mr. G was less irritable, although sexually inappropriate
behaviors and occasional episodes of yelling out (at least once a week) continued. After
four weeks, his dose of sertraline was increased to 37.5 mg daily for seven days and then
50 mg daily in the morning. Mr. G, once again, developed transient mild nausea and
anxiety, which cleared up spontaneously over one week.
After six more weeks of treatment, staff reported that sexually inappropriate behavior
had decreased considerably and episodes of yelling out now occurred only once a
month. Mr. G seemed to be getting along better with other residents and was enjoying
83
84 Mood Disorders
day-to-day activities more. His wife was also pleased to report that Mr. G was acting
much more like himself with the new medication.
Teaching Points
The selection of initial antidepressants in those requiring treatment has several potential
implications for the outcomes. The STAR*D trial showed that the rate of response to
initial antidepressant treatment is only 49.6% [1]. Another review indicated that those
who failed to respond to the first antidepressant had a 15% likelihood of suicide ideation
compared to 6% of patients with treatment-responsive depression and 1% in the general
population [2]. The financial costs of treatment for those with depression who do not
respond to the first course of antidepressants are also higher. The costs of treatment are
estimated to be $10,000/year more than for those that do respond to the initial anti-
depressant choice. Patients also vary widely in developing side effects to common
psychotropic medications. A moderate or severe side effect to an initial choice of
antidepressant can negatively impact a patient’s or family’s likelihood of pursuing
further treatment and can place a newly-formed therapeutic relationship on shaky
grounds. A study in 2014 by Hampton et al. stated that 25,000 patients in the US go to
emergency departments each year due to antidepressant-induced adverse events [3].
A major influence on positive or negative therapeutic responses to a medication is the
drug’s metabolism in a given individual, which can be hard to predict. Those familiar
with the care of older adults know that the metabolism of pharmaceuticals is influenced
by age-related factors. The liver plays a crucial role in drug metabolism and undergoes
structural and functional changes with age, including reduced blood flow and decreased
overall liver mass. These changes can lead to decreased metabolic capacity and slower
drug metabolism. Drugs are often metabolized by the cytochrome P450 microsomal
enzymes in the liver. The activity of these enzymes may decline with age, which will
prolong the rate at which some drugs are broken down. Kidney function also generally
declines with advancing age, which overall tends to lead to higher concentrations of
medications and their metabolites in older adults. Polypharmacy is also more common
in older adults and can up- or down-regulate the metabolism of pharmaceuticals and
thereby influence therapeutic effects.
How is a provider to know the likely response of a patient to a given medication when
there are so many variables? The therapeutic effects of medications are most predictable
at particular therapeutic serum concentrations, which are established during the devel-
opment and testing of a pharmacologic agent. The efficacy of neuropsychiatric medica-
tions must also consider the relationship of serum to CSF levels of drugs, which are very
dependent on the permeability of the blood–brain barrier to a given medication.
A possible, but not always reasonable, option to assess therapeutic response (or lack
thereof ) in a given patient is to measure serum drug levels. The cost of these lab tests can
be prohibitive. For example, the estimated cost for a fluoxetine serum level is estimated
at $50–200. The cost of an olanzapine serum level is estimated at $100–300 or more. This
is more than the cost of a year’s supply of this medication and insurance may or may not
cover the cost of these tests. Additionally, serum levels of fluoxetine or olanzapine are not
reliable and do not directly correlate with clinical efficacy or toxicity. Despite this, it is
still important to obtain serum drug levels to manage certain psychiatric conditions and
is a standard of care. It is also important to obtain lithium, valproic acid, and
“I Feel Very Lonely” 85
carbamazepine levels periodically when these drugs are used in the management of
bipolar disorder and some other psychiatric and neurologic conditions. Some clinicians
also utilize clozapine levels in the management of treatment-resistant schizophrenia.
Pharmacogenetic testing theoretically has the potential to increase the success rates of
initial medication selection in the treatment of depression and other psychiatric dis-
orders. The cost of this testing is significant and needs to be considered, with current
estimates around $200–1000. The costs have come down considerably in recent
years and the reports generated by these tests will list many commonly prescribed
medications from a variety of drug classes. They will estimate a patient’s likelihood to
be a normal, under, or hyper-metabolizer of a medication. Please see reference [4] for a
sample report.
There are several factors to consider before widely implementing psychopharmaco-
genetic testing in the care of psychiatric patients (Box 17.1), and several benefits may
influence a clinician’s decision to order psychiatric pharmacogenetic testing (Box 17.2)
Selective serotonin reuptake inhibitors (such as sertraline) are still considered good
first-choice antidepressants for people who have vascular disease (cardiovascular and
cerebrovascular disease) because of negligible cardiac toxicity and anticholinergic prop-
erties. Paroxetine carries a small but significant anticholinergic effect and thus is not the
SSRI of first choice for long-term care residents. Fluoxetine and fluvoxamine are long-
acting SSRIs with a significant risk of drug–drug interaction and hence are also not
first-line SSRIs for long-term care populations. Selective serotonin reuptake inhibitors
are also good first-choice antidepressants for people who have depression coexisting with
sexually inappropriate behavior, as serotonin dysfunction is associated with impulse
control disorders. Education of the family and staff in the management of adverse effects
is also key to a successful outcome.
Take-Home Points
(2017). Clinical pharmacogenetics Maciel, A., Cullors, A., Lukowiak, A. A., &
implementation consortium guideline Garces, J. (2018). Estimating cost savings of
(CPIC®) for CYP2D6 and CYP2C19 pharmacogenetic testing for depression in
genotypes and dosing of tricyclic real-world clinical settings.
antidepressants: 2016 update. Clinical Neuropsychiatric Disease and Treatment,
Pharmacology and Therapeutics, 102 (1), 37. 14, 225–230.
Section 2 Mood Disorders
18 Bipolar Disorder
Mrs. F, an 85-year-old resident, was admitted to a nursing home for rehabilitation after
hip fracture surgery. She had a long history of bipolar disorder type I with multiple
suicide attempts and hospitalizations but was stable for 10 years, taking olanzapine 5 mg
at bedtime and divalproex extended-release 500 mg twice daily. The olanzapine was
discontinued due to her experiencing “lethargy” and not participating in physical
therapy. Her daughter strongly opposed this intervention, and after Mrs. F started
showing a recurrence of bipolar depression symptoms (frequent tearfulness, loss of
appetite), her daughter insisted that olanzapine be restarted. The staff decided to seek
input from the consulting psychiatrist.
The psychiatrist obtained input from Mrs. F’s daughter, who was her primary
caregiver and had helped her through previous bouts of exacerbation of bipolar illness.
The psychiatrist felt the daughter was knowledgeable about bipolar disorder and her
mother’s treatment history. The daughter was concerned about the discontinuation of
olanzapine due to her history of severe depression and serious suicide attempts. Mrs. F’s
valproate levels were 45. During the interview, Mrs. F told the psychiatrist, “I don’t feel
right.” Upon reviewing Mrs. F’s medications, the psychiatrist noted that she was pre-
scribed oxycodone (5 mg) as needed for pain and it was being used three to four times
daily. Laboratory tests indicated mild dehydration (BUN/creatinine ratio 30 [20 and
above suggests dehydration]).
The psychiatrist recommended trying up to 3 grams/day of scheduled acetamino-
phen, encouraging intake of at least 1000 cc fluids daily, restarting olanzapine 5 mg at
bedtime, and using low-dose (2.5 mg) oxycodone as needed for breakthrough pain. Over
the next four weeks, as the use of oxycodone decreased to once a day and dehydration
resolved, the patient’s “lethargy” cleared up, her depressed mood lifted, her appetite and
sleep improved, and she started actively taking part in physical therapy.
Teaching Points
Bipolar disorder is a condition that is commonly encountered in the older adult
population and estimates are that up to 4.5% of adults in the US are affected. The
estimates for older adults are between 0.5 and 1%. Bipolar disorder usually presents early
in life with the most common diagnosis age being late adolescence or early adulthood.
There is good evidence to support bipolar disorder as being strongly influenced by
genetic factors. Monozygotic twins show a 40–70% concordance rate compared to
5–20% in dizygotic twins. Other studies have estimated the heritability of bipolar
disorder to be around 60–80%. Despite the strong influence of genetic factors on the
88
“I Don’t Feel Right” 89
Box 18.2 Commonly prescribed agents for maintenance of bipolar disorder and
monitoring issues
Mood Stabilizers:
1. Lamotrigine. The dose should be titrated slowly due to the potential for serious skin
reactions. May have antidepressant effects. Preferred by some as the first line in
older adults.
2. Divalproex. Monitor serum levels periodically. Monitor CBC periodically as divalproex
can cause thrombocytopenia or leukopenia. Monitor liver function periodically.
3. Lithium. Monitor lithium levels periodically and during acute illness. Monitor thyroid
levels and renal function periodically. A minority of patients can experience diabetes
insipidus. Generally not favored therapy in older adults due to increased risk of side
effects but may be continued if well tolerated and the patient is stable.
Second-generation antipsychotics approved for bipolar mania. Effective but be aware of
metabolic side effects. Can produce EPS side effects.
1. Olanzapine
2. Quetiapine
3. Risperidone
4. Ziprasidone
Third-generation antipsychotics approved for bipolar mania. Less likely to produce meta-
bolic side effects. Less likely to produce EPS side effects.
1. Aripiprazole
2. Asenapine
3. Cariprazine
can also be more complicated and may benefit from the input of a geriatric psychiatrist
if available.
Special consideration should also be given to those with bipolar mania or depression
with psychotic symptoms. These patients are at particularly high risk of further behav-
ioral destabilization and poor outcomes and generally require the assessment and
treatment coordinated by a psychiatrist for the best chance of stabilization and recovery.
Electroconvulsive therapy has been shown to be particularly helpful in these cases with
improvements in symptoms as soon as one or two treatment sessions.
It is also important for healthcare providers in long-term care settings to recognize
early signs of psychiatric destabilization in those with bipolar disorder. Signs of desta-
bilization in older adults can be a decreased need for sleep, increased irritability, a
general increase in activity, or even the development of psychosis (delusions or hallucin-
ations). The decompensation that can be seen in bipolar patients can happen acutely or
subacutely. The clinician should inquire about possible triggers for psychiatric destabil-
ization in bipolar patients. Possible triggers might include changes in medication adher-
ence, recent stressful life events such as the death of a loved one or changes in health
conditions, new sleep disturbances, hormonal changes (thyroid), acute medical illness,
and disruptions to the day-to-day routine. All of these factors can be implicated in
triggering worsening control of symptoms in those with bipolar disorder. Family
members can be valuable resources when evaluating changes in behavior. They have
often had many years of experience in recognizing early signs of bipolar mania or
depression in a loved one.
“I Don’t Feel Right” 91
A basic medical workup is important in ruling out a potential medical trigger such as
hypo- or hyperthyroidism, electrolyte imbalances, or infection. A good basic workup can
usually be done safely without emergency room evaluation and should include checks of
CBC, CMP, thyroid stimulating hormone (TSH), b12 level, mood stabilizer level when
indicated, urinalysis, and any other imaging or lab workup suggested by a physical exam.
Emergency room evaluation is preferred when new neurologic findings accompany
behavioral changes, any suspicion of head injury, or in those with homicidal or
suicidal behaviors.
Once underlying medical triggers for the destabilization of bipolar disorder and true
medical emergency are ruled out, the long-term care provider should quickly intervene
to prevent further worsening of bipolar symptoms and thereby prevent potential hospi-
talization. Most patients with bipolar disorder will already have mood stabilizers in place,
but the provider should assess the appropriateness of dosing. Valproic acid and lithium
levels are helpful in these instances. Therapeutic valproic acid levels are in the range of
50–100 mcg/ml. Therapeutic levels of lithium are generally considered to be
0.6–1.2 mEq/L but lower levels may be a better target for those older than 60 years of
age. Goal therapeutic levels for lithium in older adults are more appropriately
0.4–0.8 mEq/L. Doses of lithium and divalproex can be safely directed by the long-
term care provider. Lamotrigine is another commonly used mood stabilizer. Doses
effective for bipolar disorder maintenance are typically 200 mg per day in older adults.
Serum levels of lamotrigine are not commonly done or considered useful.
A commonly encountered clinical challenge is what to do if a patient on appropriate
doses of mood stabilizer and no obvious underlying medical trigger has a manic or
hypomanic episode. In these cases, a second agent may be necessary. Second-generation
antipsychotics are the most commonly used add-on options for those with mania
uncontrolled by mood stabilizers alone. Common choices include aripiprazole,
olanzapine, quetiapine, risperidone, and ziprasidone. As with other medications in older
adults, a smaller starting dose with frequent re-evaluation is preferred over starting at a
dose that is too high.
The treatment of bipolar disorder in older adults would not be complete without the
mention of nonpharmacologic interventions that can improve outcomes in those with
the condition. Psychotherapy can assist patients with bipolar disorder to self-identify
signs of mania, avoid triggers, and help patients with interpersonal conflicts that can
sometimes exacerbate or trigger symptoms of bipolar mania or depression. Lifestyle
interventions such as avoidance of stimulants like caffeine, regular physical activity,
maintaining and supporting relationships with loved ones, and regular sleep schedules
can be key to helping manage those at risk of mania or depression.
Take-Home Points
Bipolar disorder is a condition that is commonly encountered in the older adult population.
Estimates are that up to 4.5% of adults in the US are affected by bipolar disorder. The estimates
for older adults are between 0.5 and 1%.
It is important for healthcare providers in long-term care settings to recognize early signs of
psychiatric destabilization in those with bipolar disorder. Signs of destabilization in older adults
can be a decreased need for sleep, increased irritability, a general increase in activity, or even the
development of psychosis (delusions or hallucinations).
92 Mood Disorders
Starting a mood stabilizer or second-generation antipsychotic is a good first choice for those who
are depressed with a known personal history of bipolar disorder and who are not already on one.
Once underlying medical triggers for the destabilization of bipolar disorder and true medical
emergency are ruled out, the long-term care provider should quickly intervene to prevent further
worsening of bipolar symptoms and thereby prevent potential hospitalization.
Kapczinski, F., McIntyre, R. S., Kozicky, J., and International Society for Bipolar
Kanba, S., Lafer, B., Suppes, T., Calabrese, Disorders (ISBD) 2018 guidelines for the
J. R., Vieta, E., Malhi, G., Post, R. M., & management of patients with bipolar
Berk, M. (2018). Canadian Network for disorder. Bipolar Disorders, 20 (2), 97–170.
Mood and Anxiety Treatments (CANMAT) doi:10.1111/bdi.12609
Section 2 Mood Disorders
“I Am Feeling Great”
Case
19 Secondary Mania
Mr. L was a 62-year-old male who was recently transferred back to his long-term care
facility after being treated for severe pneumonia in hospital. While hospitalized, he was
diagnosed with bipolar disorder and was treated with haloperidol and lorazepam for
aggressive behaviors. Once transferred back to the long-term care facility, Mr. L was
experiencing falls and was described as being belligerent with the staff. The staff believed
he was having mood swings and exhibiting progressively worse aggressive verbal and
physical behaviors over the one to two months since being hospitalized for pneumonia.
He was also frequently irritable.
For some years before being admitted to the long-term care facility, Mr. L had a history
of making rude comments during social situations and becoming verbally abusive toward
the people he was working with. He became increasingly difficult to live with and, as a
result, his wife of 32 years filed for divorce. Mr. L’s family described an episode where he
had assaulted a friend at a friend’s house and then decided to walk two miles home in the
bitter cold. Consequently, he developed pneumonia and had to be hospitalized.
He had also become nonadherent with treatment for his diabetes, congestive heart
failure, and multiple other medical conditions. As a result, he required two partial foot
amputations and needed assistance with multiple ADLs and medication management.
This led his family to pursue placement in long-term care.
Up until about ten years ago, Mr. L was described as congenial and easy-going
throughout his adult life. Before his 50s, he had no history of psychiatric problems.
His brother was diagnosed with FTD at age 58.
During the mental status exam, Mr. L reported feeling great. He had recently
physically assaulted an acquaintance at the long-term care facility after they declined
to invest money in a real estate scheme proposed by Mr. L. When asked about his
actions, Mr. L stated that his friend deserved to be hit because he had declined Mr. L’s
offer to “make money.” Mr. L’s SLUMS score was 25. On the neurological exam, he
exhibited mild extrapyramidal symptoms, including tremors, stiffness, and bradykinesia.
Mr. L agreed to undergo neuropsychological testing and was found to have signifi-
cant executive dysfunction. An MRI of the brain revealed moderate to severe asymmetric
atrophy of both the frontal lobes and insula. The healthcare provider diagnosed him with
FTD with secondary mania.
Treatment for Mr. L was initiated with divalproex and quetiapine, while haloperidol
was discontinued. Lorazepam was switched from scheduled dosing to as-needed dosing.
Over the next four weeks, Mr. L’s aggression significantly decreased, and his behavior
became more manageable for the staff. The haloperidol-induced extrapyramidal symp-
toms completely resolved.
94
“I Am Feeling Great” 95
Teaching Points
Mania is most commonly thought of as a phase of bipolar disorder and, for this reason, it
can be easily misdiagnosed as such when a secondary cause of mania may truly be the
culprit. Primary mania results from bipolar disorder. Secondary mania is a distinct form
of mania that arises due to an underlying cause or condition. The symptoms of
secondary mania can be indistinguishable from mania associated with bipolar disorder
and can include an abnormally elevated or irritable mood accompanied by increased
energy, racing thoughts, decreased need for sleep, impulsivity, or other manic features.
Mania can occur secondary to metabolic disturbances, infection, neoplasm, epilepsy, and
medications. When mania is a symptom of an underlying medical problem it is best
considered secondary mania.
Although bipolar disorder can be seen with onset in persons over the age of
55 without a previous psychiatric history, new onset mania in older adults is more
commonly secondary to another condition. Mania in older adults can be more debilitat-
ing than in younger patients. It is also known to be associated with more cognitive
impairment in older adults.
Mania secondary to an underlying medical condition can result from various causes.
Conditions to keep in mind include primary neurological disorders, endocrine abnor-
malities, medications, illicit substances, infectious disease, metabolic abnormalities,
autoimmune disorders, and primary brain lesions (Box 19.1).
The workup of suspected secondary mania should first include a good history and
physical. The history should focus on current medical symptoms, recent infections, use
of medication or abuse of drugs, and any personal or family history of psychiatric
conditions. If any neurological findings are demonstrated on a physical exam, a neuro-
logical consultation may be helpful. Lab evaluation should include blood counts, basic
chemistries, vitamin B12, folate, and TSH levels. Imaging may include a CT of the head
or MRI depending on the clinical scenario and acuity of the situation.
The treatment of secondary mania primarily focuses on correcting or optimizing the
treatment of the underlying medical condition. There are several good options for
treating the manic symptoms themselves. Divalproex is well tolerated in most older
adults and is prescribed preferentially over valproic acid as it has much less likelihood of
GI side effects. Second-generation antipsychotics, including olanzapine, risperidone, or
quetiapine, are also good options and can be titrated easily to achieve the desired
Box 19.2 Steps in the treatment of secondary mania in the long-term care setting
1. Assess the appropriate treatment setting. Consider inpatient treatment if the patient is
a potential harm to self or others or has acute medical comorbidities.
2. Optimize treatment of the underlying medical disorder.
3. Discontinue caffeine and all alcohol if applicable.
4. Discontinue antidepressants.
5. Initiate a first-line agent such as divalproex or an atypical antipsychotic. Titrate the
dose to a therapeutic dose.
6. If not responding adequately to a first-line agent consider adding another agent. Add
divalproex if already on an atypical antipsychotic or add an atypical antipsychotic if the
patient is already on divalproex.
7. If symptoms are still not well controlled consider referral to a psychiatrist or
geriatric psychiatrist.
Take-Home Points
Mania is most commonly thought of as a phase of bipolar disorder and, for this reason, it can be
easily misdiagnosed as such when a secondary cause of mania may truly be the culprit.
The workup of suspected secondary mania should first include a good history and physical. The
history should focus on current medical symptoms, recent infections, use of medication or abuse of
drugs, and any personal or family history of psychiatric conditions.
Mania secondary to an underlying medical condition can result from various causes. Conditions to keep
in mind include primary neurological disorders, endocrine abnormalities, medications, illicit sub-
stances, infectious disease, metabolic abnormalities, autoimmune disorders, and primary brain lesions.
The treatment of secondary mania primarily focuses on correcting or optimizing the treatment of
the underlying medical condition. There are several good options for treating the manic symptoms
themselves. Divalproex is well tolerated in most older adults.
“I Am Feeling Great” 97
Mr. T, a 78-year-old divorced male, had been reporting fears of being put in jail because
he had been living “illegally” in the nursing home. He was also experiencing sleep
impairment and lack of appetite with a 15-pound weight loss over three months (from
189 pounds to 174, height was 5’ 8’’) and had been refusing to shave, change clothes, or
bathe for three weeks. He was referred to the psychiatrist because for a week he had been
saying that he would be better off dead. The psychiatrist noted that Mr. T had a history of
a “mini-stroke” six months previously and had recently developed severe abdominal pain
requiring gall bladder surgery. Mr. T was admitted to the nursing home for rehabili-
tation. After completing rehabilitation, he was transferred to the long-term care wing for
a continued stay as there was no one to take care of him at home. He also had a history of
recurrent falls, cognitive impairment, and refusal to take medication, and required close
monitoring of unstable diabetes.
His family reported that Mr. T had been disappointed when he was told he could not
return home. He had since become withdrawn, stopped smiling, and even stopped
watching sports on TV (one of his favorite activities in the past). During the interview,
Mr. T reported feeling nervous and explained that his reason for not shaving or bathing
was fear that his skin would somehow “infect” his roommate. He felt that he could not
ask for a room change because “I am staying here illegally.” Mr. T further explained that
he had no money to pay the high cost of the nursing home (he had apparently seen the
monthly charge of his stay, which was approximately $6,000). Mr. T had a history of
severe depression 15 years previously when he had retired but his daughters did not
know the details and Mr. T could not give details except to confirm that he was given
“nerve pills.” There was no obvious history of previous manic or hypomanic episodes
and Mr. T had a family history of depression (sister) and suicide (brother). Medication
review did not identify any medication that could cause or exacerbate psychiatric
symptoms, and laboratory tests (including thyroid tests and vitamin levels) were
unremarkable.
On a mental status exam, Mr. T exhibited a depressed mood and affect, delusions of
persecution, and passive suicidal ideas (felt life was not worth living) but denied any
specific suicidal intention or plan. His recall was one out of three, his SLUMS score was
16, and his geriatric depression scale (GDS30) score was 20/30 (severe depression).
Mr. T was diagnosed with recurrent MDD with psychotic features and probable vascular
MNCD. The psychiatrist recommended hospitalization and ECT, but Mr. T declined
both. There were insufficient grounds for involuntary commitment, as he agreed to
treatment at the nursing home. Mr. T was started on sertraline 25 mg daily, and the dose
was gradually increased every four days by 25 mg to 100 mg daily. He was also started on
98
“I Am Staying Here Illegally” 99
risperidone 0.25 mg at bedtime, which was increased to 0.5 mg at bedtime after one
week. Behavioral activation with an individualized pleasant activity schedule (IPAS)
was initiated.
After four weeks, family and staff reported that Mr. T’s appetite and sleep had
improved but he was still refusing to shave and come out of his room. His participation
in exercise and other activities was intermittent and brief. During the interview,
Mr. T continued to express delusional thinking and depressed affect. Over the next two
weeks, risperidone was further increased to 0.25 mg daily in the morning and 0.5 mg at
bedtime for seven days and then 0.5 mg daily in the morning and 0.5 mg daily at bedtime.
After four more weeks, Mr. T reported for the first time that he was feeling better and
started showing interest in watching sports on TV and allowed staff to shave him and
help him bathe. His participation in exercise and other pleasant activities also increased
considerably. After four more weeks, he was shaving and bathing regularly and did not
express any fears he had before. Mr. T still expressed sadness at not being able to go
home and hoped the psychiatrist would help him return. Mr. T’s repeat GDS30 score was
8/30 and his repeat SLUMS score was 21.
Teaching Points
Most clinicians treating geriatric patients are comfortable with the diagnosis of MDD.
The symptoms of major depression are listed in Box 20.1. Those with depression with
psychosis meet the criteria for diagnosis of depression but also experience psychotic
symptoms. When individuals with MDD experience delusions, hallucinations, or cata-
tonic symptoms, it is referred to as MDD with psychotic psychosis, also known as
psychotic depression. These individuals often display inflexible thinking, more severe
depression, and delusions that typically align with their mood. These delusions may
involve guilt regarding past events, poverty, somatic illness (such as an obsessive fear of
having a terminal illness like cancer), or paranoia. Hallucinations, although less common
than delusions in these cases, are more likely to be auditory rather than visual or somatic.
Residents diagnosed with psychotic depression often exhibit agitation, characterized
by classic behaviors like pacing and wringing hands. They also experience more pro-
nounced cognitive disturbances and have a higher likelihood of having a family history
of MDD or bipolar disorder. If a resident with depression has a history of delusions
during a depressive episode or shows profound anxiety, it strongly indicates the possi-
bility of psychotic depression.
The lifetime prevalence of psychotic depression has been estimated between 0.35–1%
[1]. In contrast to schizophrenia and schizoaffective disorder, the psychotic symptoms in
psychotic depression remit as the depression improves. Psychosis in bipolar disorder is
more likely to emerge during a manic episode. The severity of depression is often greater
in those with psychotic depression when compared to patients with depression without
psychosis.
The nature of psychosis in those with depression is usually mood-congruent somatic,
pessimistic, or guilt-related delusions. An example of a mood-congruent somatic delu-
sion might be a patient’s fixed belief that they are dying of a malignancy when there is no
evidence to support that belief. Delusions of spousal infidelity are also commonly seen.
Healthcare providers must diagnose psychotic depression early due to its high risk
of suicide and poor response to antidepressant treatment alone [2]. Additional
100 Mood Disorders
Additional symptoms
1. Lack of appetite or weight loss; increased appetite or weight gain.
2. Insomnia or hypersomnia.
3. Recurrent thoughts of suicide, wish to die, or suicide attempt.
4. Decreased energy or fatigue.
5. Hopelessness.
6. Helplessness.
7. Worthlessness.
8. Inappropriate or excessive guilt.
9. Loss of self-confidence.
10. Staying in bed excessively.
11. Multiple somatic complaints.
12. Chronic pain not responding to pain medication.
13. Hypochondriasis (excessive fear that one may have or will develop a serious illness).
14. Marked symptoms of anxiety.
15. Agitation (especially by residents who have advanced MNCD).
16. Verbal aggression or verbally abusive behavior (especially by residents who have
advanced MNCD).
17. Physical aggression (especially by a resident who has advanced MNCD).
18. Decreased spontaneous speech.
19. Psychomotor retardation or agitation.
20. Refusing medication.
21. Resisting care.
22. Refusing to participate in therapy (physical, occupational, speech).
23. Sudden decline in cognition.
24. Subjective memory complaint.
pharmacotherapies and is more commonly used in those with severe depression with
suicidality, catatonia, and those with psychotic depression. Studies have shown a
response rate of 70–90% with ECT in those with severe depression. The safety of the
procedure has improved due to improvements in ECT and anesthesia techniques. Side
effects can be a temporary disorientation that may be accompanied by an anterograde or
retrograde amnesia. The amnesia usually recovers over time.
Common indications for ECT include severe depression, psychotic depression,
treatment-resistant depression, catatonia, mania, and acute suicidality. Older adults
may have an even greater response to ECT than younger patients.
Regarding the case presented here, an important consideration is whether or not the
patient can be safely treated in a long-term care setting versus an inpatient setting.
Potential reasons for inpatient psychiatric care include, but are not limited to, acute
suicidality, catatonia, severe behavioral disturbances, and refusal of oral medications.
The clinician recommended that Mr. T be treated in an inpatient setting with ECT but
the patient declined this recommendation. His psychotic depression warranted closer
follow-up. It should be noted that response to antidepressant/antipsychotic medications
can take up to 12 weeks. The improvement in the SLUMS score with treatment of
depression underscores the fact that depression is often a reversible factor in the
treatment of cognitive impairment in people who have MNCD.
Take-Home Points
Those with depression with psychosis meet the criteria for diagnosis of depression but also
experience psychotic symptoms. When individuals with MDD experience delusions, hallucinations,
or catatonic symptoms, it is referred to as MDD with psychotic features, also known as
psychotic depression.
It is crucial for healthcare providers to diagnose psychotic depression early due to its high risk of
suicide and poor response to antidepressant treatment alone. Additional antipsychotic medication
is typically necessary in addition to the antidepressant for an effective response.
The nature of psychosis in those with depression is usually mood-congruent somatic, pessimistic, or
guilt-related delusions.
Electroconvulsive therapy is more commonly used in those with severe depression with suicidality,
catatonia, and those with psychotic depression. Studies have shown a response rate of 70–90%
with ECT in those with severe depression.
on the effects of electroconvulsive therapy Rothschild, A. J., Whyte, E. M., Mah, L.,
(ECT) on depressive symptoms. Canadian Nierenberg, J., Hoptman, M. J., Davatzikos,
Journal of Psychiatry. Revue Canadienne de C., Satterthwaite, T. D., & Voineskos, A. N.
Psychiatrie, 67 (5), 351–360. https://doi.org/ (2020). Structural brain networks in
10.1177/07067437211064020 remitted psychotic depression.
Lloyd, J. R., Silverman, E. R., Kugler, J. L., & Neuropsychopharmacology, 45 (7),
Cooper, J. J. (2019). Electroconvulsive 1223–1231. https://pubmed.ncbi.nlm.nih
therapy for patients with catatonia: Current .gov/32109935/
perspectives. Neuropsychiatric Disease and Wu, Z., Su, G., Lu, W., Liu, L., Zhou, Z., &
Treatment, 16, 2191–2208. https://doi.org/ Xie, B. (2021). Clinical symptoms and their
10.2147/NDT.S231573 relationship with cognitive impairment in
Neufeld, N. H., Kaczkurkin, A. N., Sotiras, A., elderly patients with depressive disorder.
Mulsant, B. H., Dickie, E. W., Flint, A. J., Frontiers in Psychiatry, 13. https://doi.org/
Meyers, B. S., Alexopoulos, G. S., 10.3389/fpsyt.2022.1009653
Section 3 Psychotic Disorders
Ms. S was a 77-year-old resident with a known schizoaffective disorder. She was
transferred to long-term care from a local group home as she had become nonadherent
with medications and had not been eating well. She had been maintained on aripiprazole
for some time, but she decided to stop the medication as she felt it was “mind-altering”
and caused her to feel not like herself. She also stated that she felt her visual hallucin-
ations were becoming more frequent and that they were caused by and not helped by the
aripiprazole. Her most frequent hallucination was that of a man sitting in her room
silently watching her, who would usually disappear once she began speaking. She also
had feelings that others were causing her harm without even speaking or interacting with
her in any way through a sort of telepathic ability.
The patient’s aripiprazole was discontinued due to her feelings that it was causing her
harm. She was started on risperidone 0.5 mg every evening but it seemed to have little
effect on her hallucinations so it was increased to 1 mg qhs. She had no prominent mood
symptoms and was not on any antidepressant medications or medications for anxiety.
She felt the higher dose of risperidone caused her GI distress. She then developed
worsening delusions, thinking that staff were trying to poison her and that the cleaning
products used in the facility were causing her to be essentially paralyzed from the waist
down. She rarely left her room and stayed mostly in her bed.
She was sent to the emergency room for making a vague suicidal statement that
she later did not recall and continued to feel that her risperidone and the cleaning
products used in the facility were causing her lower extremity weakness. She was
started on sertraline 25 mg daily for depression which was gradually increased to
100 mg daily, and had continued paranoid delusions. For example, she thought that
her priest was telling other residents at the facility about private conversations they
had had.
Over the following year, she seemed to settle in at the long-term care facility. Her
routine was mostly solitary, leaving her room only for meals and writing mostly unin-
telligible prose in a tattered notebook. She again began to decline all medications,
including her psychotropics. Despite her nonadherence to medications, she did not
develop any disruptive behaviors or worsening mood symptoms. It was decided not to
push the issue of medication nonadherence with the patient as she was functioning fairly
well. Her routine was unfaltering. She left her room only for meals and preferred to be
alone and not participate in group activities. She remained content to write in her
notebooks and had brief but friendly interactions with a few staff members she had
learned to trust. She remained relatively suspicious of most others.
103
104 Psychotic Disorders
Teaching Points
Ms. S’s case presented several challenges over the two to three years she was cared for by
her geriatric psychiatrist. Her paranoia regarding medications and staff made it nearly
impossible at times to treat her. This is a not an uncommon scenario when providing
mental health for those with thought disorders and prominent paranoia. Over the next
one to two years the patient settled down into her new community. Facility staff began to
get to know her and became less troubled by the occasional odd statements she made.
Although long-acting injectable medications (LAIM) were considered it was decided that
a more conservative watch-and-wait approach was acceptable as the patient’s delusions
and hallucinations were generally not troubling to her nor did they lead to behaviors that
were disruptive to staff or other residents. It is possible over time that Ms. S learned to be
very guarded when discussing her mental health and learned to minimize her symptoms
as a way of avoiding unwanted treatments. Regardless of the reason she continued to do
well in this environment with periodic brief encounters with her mental health providers
to assess behavioral and mood stability.
It is well known that, despite the availability of effective oral antipsychotics for the
treatment of schizophrenia, adherence can be low, with 40–50% of patients on anti-
psychotics for schizophrenia nonadherent to treatment. Nonadherence to treatment can
lead to increased relapse rates of serious symptoms and an overall decline in function.
Strategies discussed to increase adherence to antipsychotic treatment often involve long-
acting injectable antipsychotics (LAIs).
Some benefits of LAIs include improved adherence, reduced relapse rates, reduced
hospitalization rates, improved quality of life, and improved therapeutic outcomes. They
are typically administered once every four weeks, which eliminates the need for daily
medication adherence. A newer formulation of paliperidone can be administered intra-
muscularly once every three months. This can be especially beneficial for individuals
who struggle with adhering to a daily medication regimen. Studies have shown that LAIs
are associated with lower rates of relapse compared to oral medications. This may be due
to the consistent blood levels of the medication provided by LAIs, which can help to
maintain therapeutic levels and prevent symptoms from returning. Long-acting inject-
able antipsychotics have been shown to reduce the need for hospitalization due to
relapse. This can help individuals to maintain their daily lives and avoid disruptions in
their routines. Long-acting injectable antipsychotics can provide individuals with a
greater sense of control over their illness, as they do not have to worry about missing
doses or experiencing medication-related side effects as frequently. They can provide
more consistent therapeutic outcomes over time, as the medication is released slowly
into the bloodstream over weeks. This can lead to more stable symptom control and a
better overall treatment response.
Take-Home Points
Medication adherence is a major challenge when treating psychiatric conditions, especially condi-
tions that can involve impaired insight and judgment.
Long-acting injectable antipsychotics are best started by those with experience in their use but
continuing therapy may be managed by primary providers in areas underserved by mental
health specialists.
“That Priest Is a Gossip” 105
Conditions requiring strict adherence to antipsychotics may be helped by the use of LAIs. Some
available LAIs include aripiprazole, paliperidone, olanzapine, and risperidone.
Benefits of LAIs include better adherence when compared to oral therapies, the bypassing
of potentially unpredictable GI pharmacokinetics, and improved steady-state blood levels of
medication.
Ms. S was a 77-year-old resident with a known schizoaffective disorder. She was
transferred to long-term care from a local group home as she had become nonadherent
with medications and had not been eating well. She had been maintained on aripiprazole
for some time, but she decided to stop the medication as she felt it was “mind-altering”
and caused her to feel not like herself. She also stated that she felt her visual hallucin-
ations were becoming more frequent and that they were caused by and not helped by the
aripiprazole. Her most frequent hallucination was that of a man sitting in her room
silently watching her, who would usually disappear once she began speaking. She also
had feelings that others were causing her harm without even speaking or interacting with
her in any way through a sort of telepathic ability.
The patient’s aripiprazole was discontinued due to her feelings that it was causing her
harm. She was started on risperidone 0.5 mg every evening but it seemed to have little
effect on her hallucinations so it was increased to 1 mg qhs. She had no prominent mood
symptoms and was not on any antidepressant medications or medications for anxiety.
She felt the higher dose of risperidone caused her GI distress. She then developed
worsening delusions, thinking that staff were trying to poison her and that the cleaning
products used in the facility were causing her to be essentially paralyzed from the waist
down. She rarely left her room and stayed mostly in her bed.
She was sent to the emergency room for making a vague suicidal statement that
she later did not recall and continued to feel that her risperidone and the cleaning
products used in the facility were causing her lower extremity weakness. She was
started on sertraline 25 mg daily for depression which was gradually increased to
100 mg daily, and had continued paranoid delusions. For example, she thought that
her priest was telling other residents at the facility about private conversations they
had had.
Over the following year, she seemed to settle in at the long-term care facility. Her
routine was mostly solitary, leaving her room only for meals and writing mostly unin-
telligible prose in a tattered notebook. She again began to decline all medications,
including her psychotropics. Despite her nonadherence to medications, she did not
develop any disruptive behaviors or worsening mood symptoms. It was decided not to
push the issue of medication nonadherence with the patient as she was functioning fairly
well. Her routine was unfaltering. She left her room only for meals and preferred to be
alone and not participate in group activities. She remained content to write in her
notebooks and had brief but friendly interactions with a few staff members she had
learned to trust. She remained relatively suspicious of most others.
103
104 Psychotic Disorders
Teaching Points
Ms. S’s case presented several challenges over the two to three years she was cared for by
her geriatric psychiatrist. Her paranoia regarding medications and staff made it nearly
impossible at times to treat her. This is a not an uncommon scenario when providing
mental health for those with thought disorders and prominent paranoia. Over the next
one to two years the patient settled down into her new community. Facility staff began to
get to know her and became less troubled by the occasional odd statements she made.
Although long-acting injectable medications (LAIM) were considered it was decided that
a more conservative watch-and-wait approach was acceptable as the patient’s delusions
and hallucinations were generally not troubling to her nor did they lead to behaviors that
were disruptive to staff or other residents. It is possible over time that Ms. S learned to be
very guarded when discussing her mental health and learned to minimize her symptoms
as a way of avoiding unwanted treatments. Regardless of the reason she continued to do
well in this environment with periodic brief encounters with her mental health providers
to assess behavioral and mood stability.
It is well known that, despite the availability of effective oral antipsychotics for the
treatment of schizophrenia, adherence can be low, with 40–50% of patients on anti-
psychotics for schizophrenia nonadherent to treatment. Nonadherence to treatment can
lead to increased relapse rates of serious symptoms and an overall decline in function.
Strategies discussed to increase adherence to antipsychotic treatment often involve long-
acting injectable antipsychotics (LAIs).
Some benefits of LAIs include improved adherence, reduced relapse rates, reduced
hospitalization rates, improved quality of life, and improved therapeutic outcomes. They
are typically administered once every four weeks, which eliminates the need for daily
medication adherence. A newer formulation of paliperidone can be administered intra-
muscularly once every three months. This can be especially beneficial for individuals
who struggle with adhering to a daily medication regimen. Studies have shown that LAIs
are associated with lower rates of relapse compared to oral medications. This may be due
to the consistent blood levels of the medication provided by LAIs, which can help to
maintain therapeutic levels and prevent symptoms from returning. Long-acting inject-
able antipsychotics have been shown to reduce the need for hospitalization due to
relapse. This can help individuals to maintain their daily lives and avoid disruptions in
their routines. Long-acting injectable antipsychotics can provide individuals with a
greater sense of control over their illness, as they do not have to worry about missing
doses or experiencing medication-related side effects as frequently. They can provide
more consistent therapeutic outcomes over time, as the medication is released slowly
into the bloodstream over weeks. This can lead to more stable symptom control and a
better overall treatment response.
Take-Home Points
Medication adherence is a major challenge when treating psychiatric conditions, especially condi-
tions that can involve impaired insight and judgment.
Long-acting injectable antipsychotics are best started by those with experience in their use but
continuing therapy may be managed by primary providers in areas underserved by mental
health specialists.
“That Priest Is a Gossip” 105
Conditions requiring strict adherence to antipsychotics may be helped by the use of LAIs. Some
available LAIs include aripiprazole, paliperidone, olanzapine, and risperidone.
Benefits of LAIs include better adherence when compared to oral therapies, the bypassing
of potentially unpredictable GI pharmacokinetics, and improved steady-state blood levels of
medication.
22 Catatonia
Mr. S was a 68-year-old male with a history of congestive heart failure, diabetes, morbid
obesity, frequent UTIs, bipolar disorder, benign prostatic hypertrophy, urinary reten-
tion, remote history of deep vein thrombosis (DVT), and a history of decubitus ulcers.
He had lived in long-term care for the past five years as his physical disability from his
congestive heart failure (CHF) and obesity progressed. He required a motorized wheel-
chair for mobility and was dependent on caregivers for nearly all ADLs. He had a
supportive family, his wife and two daughters lived close to his care facility and were
able to visit often.
Mr. S had been followed by his long-term care psychiatrist for four years. He was
doing well on his current regimen of divalproex 500 mg bid and citalopram 20 mg daily.
His non-psychiatric medications included tamsulosin 0.4 mg daily, mirabegron 25 mg
daily for an overactive bladder, carvedilol 12.5 mg twice daily, fosinopril 10 mg daily,
torsemide 20 mg daily, spironolactone 25 mg daily, glipizide 5 mg daily, and aspirin
81 mg daily.
Unfortunately, Mr. S had an injury while being transferred from his bed to his
wheelchair and sustained a fracture to his left hip requiring admission to the hospital
for an open reduction-internal fixation procedure. He was hospitalized for four days and
discharged back to the nursing home. After coming back to the nursing home the patient
was noted to be more withdrawn than usual. Facility care staff alerted his psychiatrist
that Mr. S was often found in his wheelchair just staring into space and found little
interest in the social activities at the facility that he previously enjoyed. His wife also
noted that on her most recent visit, Mr. S was inattentive and did not appear to enjoy her
visit, which was quite unlike him. Staff also mentioned that Mr. S preferred to stay in his
room in the mornings and often skipped breakfast and sometimes lunch.
On examination, the patient was found to be appropriately dressed and he was in his
wheelchair as usual. Eye contact was poor, his mood was described as depressed, and his
affect was flat. There was no evidence of hallucinations or delusions and Mr. S would
generally respond to questions with only brief one-word responses. His psychiatrist
diagnosed worsening depression in the setting of bipolar disease and added aripiprazole
2.5 mg, which was later increased to 5 mg daily. At a return visit the following week, the
patient had not improved and continued to spend most of the day in bed. He was eating
and drinking little and only with much encouragement. It was thought that Mr. S’s
depression had not had time enough to respond to the addition of aripiprazole. Labs
including a urinalysis were recommended but no acute changes to explain his worsening
lethargy were found.
106
“He Just Won’t Get Up” 107
One of the members of staff at the facility later reached out to the psychiatrist and
said that the patient’s status had again worsened, and he was now refusing to get out of
bed. He was in bed nearly all of the time with his eyes closed and responding minimally
to others. He had no interest in even watching television. He was now eating only bites of
nutrition at best in a given day. He was not opening his mouth for medications.
An urgent visit by a psychiatrist was requested.
At the follow-up visit Mr. S was found to be in his bed with his eyes closed at 1 pm.
His nurse commented, “he just won’t get up.” His hygiene was poor and he was
disheveled. When addressed verbally he would not answer questions and would only
briefly open his eyes when shaken. When asked how he felt he would reply with the word
“fine”, whispered several times. His limbs were described as somewhat stiff. Mr. S’s wife
was called and permitted the patient to be transferred to a hospital with geriatric
psychiatric services for further workup and treatment of presumed catatonia. Mr. S’s
workup in the emergency department included imaging and labs that did not show any
anatomic or metabolic causes for his worsening mental status. An inpatient psychiatric
evaluation was obtained and the impression was that of catatonia in the setting of bipolar
depression. Psychiatry recommended starting lorazepam at 0.5 mg IV three times daily.
Within 24 hours Mr. S became somewhat more alert and responsive, as demonstrated by
making eye contact with his family and nursing staff and answering simple yes/no
questions appropriately. The dose of lorazepam was increased to 1 mg three times daily.
Over the next two days, Mr. S became increasingly alert and began to take in small
amounts of food and fluids. Soon he was able to maintain his hydration without IV fluids
and was transferred to a geriatric psychiatric unit for further treatment.
Teaching Points
Catatonia is a severe neuropsychiatric condition characterized by a state of immobility,
stupor, and unresponsiveness to the environment. In years past the diagnosis of catatonia
implied the presence of underlying schizophrenia but now it is known to be associated
with a variety of psychiatric, neurological, or medical disorders. It is usually seen in
mood disorders but can also be seen in psychotic and various neurological disorders.
Estimates vary depending on location, but it is thought to be present in 5–10% of
inpatient psychiatric patients.
The symptoms of catatonia are listed in Box 22.1. Signs and symptoms of catatonia can
be thought of in terms of motor signs, affective features, and cognitive–behavioral features.
Motor signs may include posturing, waxy flexibility, stereotypy, and dyskinesias. Examples
of affective features of catatonia can include emotional lability, anxiety, affective latency,
impulsivity, and excitement. Cognitive–behavioral features can include such signs as
perseveration, echolalia, echopraxia, mutism, and automatic obedience. Rating scales
such as the Bush–Francis Catatonia Rating Scale can be used to assist in the evaluation
of patients with suspected catatonia [1]. The Bush–Francis rating scale helps
clinicians quantify the severity of motor signs, affective signs, and cognitive–behavioral
features.
The causes of catatonia can vary and include psychiatric disorders such as schizo-
phrenia, bipolar disorder, and MDD, as well as medical conditions such as encephalitis,
brain injury, and metabolic disorders. See Box 22.2 for a partial list of medical conditions
which can be accompanied by catatonic features.
The pathophysiology of catatonia is not fully understood, but it is believed to involve
dysfunction in the dopamine and gamma-aminobutyric acid (GABA) neurotransmitter
systems, as well as abnormalities in the frontal–subcortical circuits and the limbic
system.
Some studies suggest that catatonia may be related to changes in the balance of
excitatory and inhibitory neurotransmitters in the brain. Specifically, it is thought that
decreased GABA activity in the frontal lobes may lead to overstimulation of the basal
ganglia and other subcortical regions, resulting in the characteristic motor abnormalities
seen in catatonia.
Other studies have implicated abnormalities in the dopamine system, which may
contribute to the motor symptoms and cognitive deficits seen in catatonia. For example,
dopamine receptor hypersensitivity has been observed in some cases of catatonia, which
may explain the hyperactivity and agitation seen in some patients.
In addition to these neurotransmitter abnormalities, catatonia has also been associ-
ated with changes in brain structure and function. Some studies have suggested that
“He Just Won’t Get Up” 109
patients with catatonia may have reduced gray matter volume in certain brain regions
including the anterior cingulate cortex and the prefrontal cortex. Other studies have
found alterations in functional connectivity between brain regions, particularly in the
frontal–subcortical circuits [2].
The pathophysiology of catatonia is complex and likely involves dysfunction in
multiple brain regions and neurotransmitter systems. Further research is needed to fully
understand the underlying mechanisms of this syndrome and to develop more
effective treatments.
The workup of suspected catatonia is in line with the workup of change in mental
status and usually includes basic labs including thyroid function, CBC, CMP, urinalysis,
and drug screen. Central nervous system imaging is usually helpful in ruling out space-
occupying lesions, stroke, and inflammatory CNS findings. An electroencephalogram
(EEG) can be necessary to rule out an underlying seizure disorder. After underlying
medical conditions have been ruled out, a benzodiazepine challenge test can help
diagnose catatonia. First, the examiner evaluates catatonic symptoms using a catatonia
rating scale such as the Bush–Francis Catatonia Rating Scale [3]. Then a dose of IV
lorazepam is given and the rating scale is repeated. A 50% reduction in the rating scale
score is very suggestive of catatonia.
The treatment of catatonia associated with medical illness is primarily the treatment
of the underlying disease. In those with catatonia secondary to psychiatric illness,
benzodiazepines and ECT are commonly used to treat catatonia. Both are effective in
reducing symptoms. Other treatments that have been used with some success include
atypical antipsychotics, such as clozapine and olanzapine, and dopamine agonists, such
as amantadine.
Benzodiazepines and ECT are the most commonly used treatments for catatonia and
both have shown similar efficacy [4]. A benzodiazepine challenge is often a good place to
start when initiating treatment for catatonia secondary to underlying psychiatric illness.
Lorazepam is the most commonly used benzodiazepine for this indication. In older
adults, a common starting dose is 0.5–1 mg IV three times daily, although higher doses
may be required. Electroconvulsive therapy can produce the highest success in cases of
catatonia [5], but does present some challenges as a treatment option. There may be
limited availability of ECT in some settings or delays in accessing it. There are also
potential legal issues in obtaining consent for ECT as patients are often not able to
consent for themselves.
Overall, the prognosis for individuals with catatonia is generally good, especially
when the underlying cause is identified and treated promptly. With appropriate treat-
ment, most individuals with catatonia will recover fully or experience significant
improvement in their symptoms.
Take-Home Points
Signs and symptoms of catatonia can be thought of in terms of motor signs, affective features, and
cognitive–behavioral features.
Benzodiazepines and ECT are the most commonly used treatments for catatonia. Both treatments
have shown similar efficacy.
23 by a Fungus!”
Delusional Disorder
Mrs. S, an 83-year-old long-term care resident, was referred to psychiatry by the patient’s
primary care nurse practitioner at the request of the patient’s daughters due to persever-
ation regarding thoughts that her body was being taken over by a fungal infection.
During the first visit, the patient’s two daughters were present and assisted with the
history. The patient insisted on providing a detailed history of a fungal infection that she
claimed started several months earlier as flakiness of her scalp. She was prescribed
Nizoral shampoo, which proved to be effective. About a month later, the patient
developed a mild candidal rash under her breasts, which also responded well to topical
Nystatin cream.
Over the subsequent months, the patient began to perseverate more about her belief
that the fungal infection was now “inside me” and “eating me from the inside out!”
When asked about the evidence that led her to believe she had a fungal infection,
Mrs. S would repeatedly respond, “I just know. I can feel it. It makes me short of breath,
I itch, I can’t breathe well. It’s in my bowels, and I can hear it making noises in there!”
The patient’s daughters advised that the patient had seen two different gastroenter-
ologists, a dermatologist, and a pulmonologist for these complaints. All of these pro-
viders released her from their care after complete workups that were negative for any
signs of systemic infection or any other acute pathology, for that matter. Despite these
reassurances, Mrs. S would fixate on perceived irregularities in her skin coloration,
breathing patterns, and bowel movements, which she was sure proved her systemic
fungal infection. These thoughts were intrusive and present every day for many hours.
Mrs. S denied having any visual or auditory hallucinations and her mood seemed
somewhat more irritable in recent weeks. Her daughters felt this irritability was second-
ary to Mrs. S’s frustration about how she felt no one was taking her claims of
infection seriously.
Mrs. S denied excessive anger, sleep difficulties, changes in appetite, or major weight
changes in the previous months.
Her psychiatric history was significant for being treated for anxiety most of her adult
life. Her daughters felt that her anxiety had been very well controlled for years with
escitalopram 10 mg daily and trazodone 25 mg at bedtime for sleep. She had never been
hospitalized for any mental health reasons. She had never been under the care of a
psychiatrist in the past and seemed offended when introduced to a psychiatrist. “I don’t
need a shrink! My mind is fine. I need someone to tell me what’s wrong with my body!”
She had been in long-term care for five to six years after her husband died suddenly.
He was instrumental in her care before his death as Mrs. S needed a fair amount of
assistance with ADLs secondary to a history of chronic back pain and the effects of
111
112 Psychotic Disorders
multiple back surgeries over the years. She had adjusted quickly and easily to life in long-
term care. She enjoyed having the assistance and companionship of care staff and had
several close friends in the facility whom she ate meals with regularly. She loved
television and could often be found watching old television shows. Her favorites were
Murder She Wrote and The Golden Girls.
On exam, Mrs. S seemed in fairly stable health. She was in a wheelchair and able to
answer all questions, and was oriented to her name, birth date, place, and year. She got a
5/5 on her serial 7s and had no deficits in her clock drawing. Her mood could be
described as euthymic to mildly anxious when discussing anything related to her
perceived fungal infection and had no evidence of any hallucinations. She was somewhat
tangential and would often bring any conversation topic back to her “fungal infection”.
When asked whether she was bothered by her consistent thoughts of her fungal
infections and her physical sensations related to them, she replied, “I’m just pissed that
no one believes me or can tell me why this is happening to me!”
At the end of the visit, the provider discreetly asked one of the daughters to help him
find his way to the front door of the facility. The patient’s daughter expressed exasper-
ation regarding the situation she and her sister found themselves in. “We don’t know
what to do with Mom . . . Every doctor has told her that it is in her head, but she is
driving us crazy with all this talk of having a fungus in her body.” The provider educated
the daughter on the condition of delusional disorder and how the disorder can cause
those afflicted to be truly convinced of their illness despite multiple medical workups
proving otherwise. The patient’s daughter was very relieved to hear that medications are
often helpful for this form of mental illness and can be well tolerated.
Mrs. S was started on 2 mg of aripiprazole qhs and maintained her low dose of
trazodone at night and her daily 10 mg dose of escitalopram, as overall it had been very
helpful with the patient’s mood and anxiety symptoms in the past. The provider returned
for a follow-up visit four weeks later and was pleased to hear that the patient was much
less troubled by her perceived illness and was spending much less time speaking of it with
her daughters and friends. When the provider sat down to speak to the patient, she
shortly began speaking of her ongoing symptoms of her perceived fungal infection. She
told the psychiatrist that the symptoms seemed to be getting a little better since the
psychiatrist started her on “that new medication” (referring to aripiprazole). The psych-
iatrist made a small increase in the dose of her aripiprazole to 5 mg qhs. At a second
follow-up visit six weeks later, the patient was in a pleasant mood. She felt nearly “cured”
of her fungal infection, and it was bothering her very little. She was again a pleasure for
her daughters to visit, as she was back to her old self and seemed little troubled by
anything, as long as she had plenty of chocolate and The Golden Girls to watch on TV.
Teaching Points
Delusional disorder is a mental illness characterized by the presence of one or more
delusions for at least one month. Delusional beliefs are based on the misinterpretation of
external reality and are not improved by education or persuasion. Major mood symp-
toms are generally absent. The delusions can result in significant distress or dysfunction
and can trouble patients, caregivers, and families alike. Delusions that involve physical
symptoms can be particularly burdensome, as the pursuit of these symptoms can lead to
unnecessary medical workup and expense.
“My Body Is Being Taken over by a Fungus!” 113
Specify whether:
Erotomanic type: This subtype applies when the central theme of the delusion is that
another person is in love with the individual.
Grandiose type: This subtype applies when the central theme of the delusion is the
conviction of having some great (but unrecognized) talent or insight or having made
some important discovery.
Jealous type: This subtype applies when the central theme of the individual’s delusion is
that his or her spouse or lover is unfaithful.
Persecutory type: This subtype applies when the central theme of the delusion involves
the individual’s belief that he or she is being conspired against, cheated, spied on,
followed, poisoned or drugged, maliciously maligned, harassed, or obstructed in the
pursuit of long-term goals.
Somatic type: This subtype applies when the central theme of the delusion involves
bodily functions or sensations.
Mixed type: This subtype applies when no one delusional theme predominates.
Unspecified type: This subtype applies when the dominant delusional belief cannot be
clearly determined or is not described in the specific types (e.g., referential delusions
without a prominent persecutory or grandiose component).
Specify if:
With bizarre content: Delusions are deemed bizarre if they are clearly implausible, not
understandable, and not derived from ordinary life experiences (e.g., an individual’s
belief that a stranger has removed his or her internal organs and replaced them with
someone else’s organs without leaving any wounds or scars).
Specify if: The following course specifiers are only to be used after a one-year duration of
the disorder:
First episode, currently in acute episode: First manifestation of the disorder meeting
the defining diagnostic symptom and time criteria. An acute episode is a time period in
which the symptom criteria are fulfilled.
First episode, currently in partial remission: Partial remission is a time period during
which an improvement after a previous episode is maintained and in which the defining
criteria of the disorder are only partially fulfilled.
First episode, currently in full remission: Full remission is a period of time after a
previous episode during which no disorder-specific symptoms are present.
“My Body Is Being Taken over by a Fungus!” 115
Unspecified
Specify current severity: Severity is rated by a quantitative assessment of the primary
symptoms of psychosis, including delusions, hallucinations, disorganized speech,
abnormal psychomotor 106 behavior, and negative symptoms. Each of these symptoms
may be rated for its current severity (most severe in the last seven days) on a five-point
scale ranging from zero (not present) to four (present and severe). (See Clinician-Rated
Dimensions of Psychosis Symptom Severity in the chapter “Assessment Measures.”)
Note: Diagnosis of delusional disorder can be made without using this severity specifier.
Take-Home Points
Delusional disorder is a mental illness characterized by the presence of one or more delusions for a
period of at least one month. Delusional beliefs are based on the misinterpretation of external
reality and are not made better with education or persuasion.
Seven subtypes of delusional disorder are recognized in the DSM–5. These include persecutory
type, somatic type, jealous type, grandiose type, erotomanic type, mixed type, and unspecified
type.
116 Psychotic Disorders
The prevalence of delusional disorder in older adults is thought to be double that seen in younger
adults. The occurrence of delusional disorder is more common in later life when compared to other
psychotic disorders such as schizophrenia.
Studies have not shown dramatic differences in response rates to different second- or third-
generation antipsychotics.
Choice of antipsychotic depends on clinician familiarity, side effect profile, and comorbidities.
“This Is Humiliating”
Case
24 Clozapine
Mr. S was a 58-year-old single male who had chronic paranoid schizophrenia and lived
in a community nursing home. He had been moved there several years before from the
state hospital ward/unit for chronically mentally ill patients. Mr. S was stable on his
antipsychotic drug regimen of clozapine 100 mg qam and 300 mg qhs. He was mostly
reclusive but did leave his room to go to the dining room for meals. Mr. S collected
pornographic magazines and videotapes, which he would read and watch in the privacy
of his room. One day, on returning from breakfast in the dining room, he found his
dresser and other belongings in the hallway. He was not told that his room would be
recarpeted. His magazines and tapes were also moved into the hallway. He became angry,
agitated, and upset – accusing the staff of messing with his personal belongings. “This is
humiliating,” he stated. His dormant suspiciousness became blatant, and he required
hospitalization after a serious suicide attempt.
Teaching Points
Knowing a resident and taking extra precautions to ensure privacy for residents who are
managing their own sexual needs appropriately is crucial, not only to support their rights
of sexual expression but also to prevent serious trauma and humiliation experienced by
residents whose privacy is violated due to lack of mindfulness on the part of staff. The
range of sexuality, romance, and sexual identity experiences in later life encompasses
various activities and experiences, including self-pleasure, physical intimacy, as well as
emotional and intellectual closeness. There is evidence to suggest that sexual expression
in later life may have positive psychological and physiological effects, such as enhancing
overall quality of life, reducing rates of depressive symptoms, and lowering the risk of
certain types of cancers and fatal coronary events [1].
Despite the well-documented benefits of continuing sexual expression in later life,
research has shown that the personal perspectives of staff and family members often
create limitations for resident sexual expression [2]. Every opportunity should be utilized
to educate all staff in long-term care about the rights of residents to privacy and
appropriate expression of sexual needs.
This case also mentions the use of clozapine. Clozapine has an important place in the
treatment of psychiatric patients. It was developed and introduced to the market in
1972 in Switzerland by the pharmaceutical company Sandoz (now Novartis). However,
in the late 1970s and early 1980s, reports emerged linking clozapine to potentially fatal
agranulocytosis, which led to the drug being withdrawn from the market in most
117
118 Psychotic Disorders
countries. After extensive research and clinical trials, clozapine was reintroduced to the
market in 1990, with additional safety measures in place to monitor for the development
of agranulocytosis. Clozapine is now an important medication in the treatment of
schizophrenia. It is used primarily in cases of treatment-resistant schizophrenia (TRS)
but is also useful in patients with schizophrenia who have had adverse effects to multiple
other antipsychotics. Treatment-resistant schizophrenia is defined as a lack of significant
improvement in symptoms despite at least two trials of different antipsychotic medica-
tions, each given at an adequate dose for an adequate duration of time. Treatment-
resistant schizophrenia can also refer to cases in which a person may respond well to an
antipsychotic medication initially but then experience a relapse of symptoms later
on, despite continuing to take the medication as prescribed [3]. Clozapine has not
proven more efficacious than other antipsychotics for the initial choice of treatment of
schizophrenia [4].
The use of clozapine has several benefits in addition to improving the control of the
symptoms of schizophrenia for those with TRS. Patients who carry the diagnosis of
schizophrenia have an elevated mortality compared to the general population, with
estimates that it is 2.5 times higher. Those with schizophrenia are also estimated to have
a lifespan that is 15–25 years shorter than those without the condition. Clozapine is not
only attractive for its benefits for those with TRS but also because it has been shown to
reduce overall mortality. It is thought to do this through a reduction in the incidence of
suicidality [5]. An additional benefit of clozapine is its documented effects and support-
ing data on the patterns of illicit substance use in those with schizophrenia [6].
Comorbid illicit substance use in schizophrenia is associated with a poor outcome,
including an increased risk of suicide and poor response to treatment. Finally, clozapine
is also not known to produce extrapyramidal symptoms that can be more common with
other first- and second-generation antipsychotics.
The risks of using clozapine are well known and the most feared side effect is
agranulocytosis. Agranulocytosis risk is highest in the first three months of use of the
drug but is possible even after years of use [4]. For this reason, most countries have a
mandatory system of lab monitoring and reporting to help minimize this compli-
cation. Some experts argue that the risk of developing agranulocytosis decreases over
time in people who use clozapine, and that ongoing monitoring may not be necessary
for those who have been stable on the medication for an extended period. However,
others argue that regular monitoring is still necessary to ensure early detection and
treatment of agranulocytosis, which can occur even after many years of clozapine use
[7]. The current standard of care is for weekly monitoring of blood counts for six
months, followed by every other week monitoring for six months, followed by
monthly monitoring thereafter. This is the main limitation of clozapine use in older
adults.
There are less common but still well-documented side effects of clozapine, which are
documented in Box 24.1.
The dosing of clozapine is not complex but warrants discussion. There are recom-
mendations to slowly titrate the dose when initiating therapy in order to minimize side
effects. It should be noted that cigarette smoking increases the metabolism of clozapine
by up to 50%. Those that suddenly stop smoking, as often happens during hospital
admission, can develop the toxic effects of clozapine in as little as several days. Nicotine
replacement therapy does not increase the metabolism of clozapine [9].
“This Is Humiliating” 119
There are some off-label uses of clozapine that healthcare providers in long-term care
should be aware of. Some studies have suggested that clozapine may be effective in
treating TRD and it has been used off-label in the treatment of bipolar disorder,
particularly in people who have not responded to other more commonly prescribed
medications. Clozapine is sometimes used in Parkinson’s disease-related psychosis and
some studies have shown that it may also be effective in treating certain symptoms of
autism spectrum disorder, such as aggression and irritability [10]. Regardless of the
indication, clozapine is probably underappreciated and underutilized in the long-term
care population.
Take-Home Points
“I Want to Go Home”
Case
25 Delirium
Mr. A was an 80-year-old male who had hypertension, benign prostatic hypertrophy,
hyperlipidemia, and arthritis. In the hospital after hip fracture surgery, Mr. A became
disoriented, insisting that he needed to go to work, tried to climb out of bed, and became
aggressive when the staff tried to redirect him. He was given haloperidol (2 mg twice
daily) and was recommended to continue haloperidol when he was transferred to a
nursing home for rehabilitation. Mr. A developed drooling, tremors, and daytime
sedation and continued to have periods of agitation. A psychiatric consultation was
requested.
The psychiatrist asked Mrs. A about Mr. A’s cognitive functioning before surgery.
Mrs. A insisted that Mr. A’s memory was “quite good” but added, “he is quite old, you
know.” Mr. A’s daughter reported that for the last two years her father had been
increasingly forgetful, repeating himself often, and growing irritable. In the last six
months he had started making mistakes in bill paying, became lost while driving, started
avoiding social gatherings, and refused to wear a hearing aid. During the examination,
the psychiatrist found Mr. A somewhat drowsy, with poor attention and concentration.
Mr. A stated to the psychiatrist, “I want to go home.” He did not recall having been
hospitalized and had slurred speech. The psychiatrist reviewed the results of tests done in
the hospital. A CT scan of the brain showed diffuse moderate cerebral atrophy, urine
analysis was negative for infection, and the results of blood tests, including the chemistry
panel, thyroid profile, and folate levels, were within normal limits. Vitamin B12 was at
the lower limit of normal (level of 220). The psychiatrist made the diagnosis of MNCD
due to probable Alzheimer’s disease with postoperative delirium, discontinued haloperi-
dol due to adverse effects caused by it, added vitamin B12 1000 mcg daily, and started
donepezil 5 mg daily in the morning to be taken after breakfast. The psychiatrist also met
with the family and staff to discuss various psychosocial environmental interventions to
be instituted (such as listening to favorite music, aromatherapy with lavender lotion,
one-to-one supervision at times, minimizing nighttime noise, and lowering expectations
for the resident to remember and follow directions). The psychiatrist then had a lengthy
family meeting at which he explained his diagnosis to the family, recommended a
support group for the wife and daughter, and discussed the prognosis.
Over the next several days, Mr. A gradually became less agitated and started taking
part in physical therapy (although he required constant reassurance, redirection, and
encouragement). The drooling was markedly reduced, tremors and slurred speech
decreased significantly, and daytime sleepiness resolved completely. After six weeks,
Mr. A was found to smile and joke, although he still asked when he could go home.
121
122 Psychotic Disorders
Teaching Points
The DSM–5–TR of the American Psychiatric Association defines delirium as, “a disturb-
ance in attention (i.e. reduced ability to direct, focus, sustain, and shift attention)
accompanied by reduced awareness of the environment” [1]. Unlike dementia, delirium
has a rapid onset and fluctuating and reversible symptoms. However, the symptoms are
unpredictable and irregular, making delirium under-detected. Delirium is categorized as
hyperactive, hypoactive, or mixed based on the predominance of psychomotor
symptoms.
Delirium is a frequently encountered syndrome in hospitalized patients, with an
estimated occurrence rate of 8–17% in older patients in emergency departments and
29–64% in general medical and older adult inpatients. This syndrome is more commonly
observed in patients with risk factors such as pre-existing cognitive impairment, recent
surgery, acute infection, or critical illness [2].
The causes of delirium are varied. The most common causes in the long-term care
setting are likely to be UTIs, untreated pain, and medication side effects (Box 25.1).
Since the etiology of delirium is multifactorial, there are probably various neurobio-
logical mechanisms that contribute to its pathogenesis, such as neuroinflammation,
brain vascular dysfunction, altered brain metabolism, neurotransmitter imbalances,
and impaired neuronal network connectivity [3]. While several neurotransmitter systems
have been associated with delirium, current literature mostly supports the theory of a
relative acetylcholine deficiency and/or dopamine excess.
Several potentially modifiable risk factors for delirium can be addressed and thereby
help prevent its development in both the hospital and long-term care settings. Modifiable
risk factors for delirium are shown in Box 25.2 [4].
Box 25.2 Potentially modifiable risk factors for delirium in older adults in the
long-term care setting
1. Medication management: Avoiding medications that can cause delirium or adjusting
doses to the lowest effective amount.
2. Dehydration and malnutrition: Addressing these issues and ensuring adequate
hydration and nutrition.
3. Infections: Identifying and treating infections promptly.
4. Pain: Managing pain effectively to reduce the risk of delirium.
5. Sleep deprivation: Encouraging good sleep habits and reducing interruptions to sleep.
6. Immobility: Encouraging activity and mobility as much as possible.
7. Sensory impairment: Addressing vision and hearing impairment.
8. Environmental factors: Reducing noise, providing natural light and orientation cues,
and encouraging family involvement and socialization.
possible. Their continued use can hinder patient progress in the long-term care setting by
causing excessive sedation, movement disorders, and worsening cognition.
When evaluating a patient in the long-term care setting with potential new-onset
delirium it is important to consider all possible causes. If symptoms of delirium are
severe, transfer to a hospital setting for timely workup and treatment is indicated.
If symptoms of delirium are mild a workup in the long-term care setting can usually
be justified and would include a urinalysis, basic labs such as a CBC, CMP, TSH, B12,
and vitamin D level, and other labs or imaging studies as indicated by history and
physical exam findings.
As in the case here, mild Alzheimer’s disease often goes unrecognized and undiagnosed.
It predisposes the person to develop postoperative delirium. Hence, for any resident who
has postoperative delirium, the healthcare provider must inquire about cognitive function-
ing before surgery. It is also common for delirium to be treated with larger doses of
haloperidol in the hospital, and patients are often discharged on haloperidol or another
antipsychotic medication. Haloperidol is associated with a high incidence of extrapyramidal
adverse effects in older adults who have MNCD, especially at doses larger than 1–2 mg/day
(although extrapyramidal symptoms [EPS] can be seen at doses as low as 0.5 mg/day).
Hence, in the management of delirium, the healthcare professional must discontinue
haloperidol or any other antipsychotic as soon as possible, especially if adverse effects arise.
Finally, besides treatment of the cause, management of delirium primarily involves
psychosocial interventions. These can include environmental modifications such as
addressing poor lighting, excessive noise, or lack of orientation cues. They should
include cognitive stimulation with activities that may include puzzles, games, and other
mentally stimulating activities to encourage engagement and interaction with the envir-
onment. Family involvement can also help. Family members can provide comfort and
support, help with orientation, and monitor the patient for changes in behavior or other
symptoms. Music therapy can be an effective nonpharmacological intervention for the
treatment of delirium. Listening to music can help promote relaxation and reduce
anxiety and agitation. Pet therapy involves the use of animals to provide comfort and
support to patients. Studies have shown that pet therapy can be effective in reducing
symptoms of delirium in some patients [7].
In summary, delirium can be seen with some frequency in the long-term care setting.
Clinicians should be aware of its potential and familiar with the treatment, workup, and
prevention.
Take-Home Points
“I Am a Worrier”
Case
Ms. S was a 69-year-old single woman who had been living in a long-term care facility for
two years due to sequelae of severe arthritis, obesity, CHF, chronic kidney disease,
chronic pain, diabetes requiring insulin, poor mobility, and osteoporosis. She had a long
history of schizoaffective disorder with multiple hospitalizations for serious suicide
attempts, severe psychotic symptoms (paranoid delusions and auditory hallucinations),
and severe depressive symptoms, but had been stable for seven years on a medication
regimen that included olanzapine 10 mg at bedtime and mirtazapine 30 mg at bedtime.
The psychiatrist was consulted for the management of increased agitation, as demon-
strated by yelling out instead of using the call light, calling her roommate “the devil”,
paranoia (accusing staff of stealing her clothes), and insomnia.
Ms. S had relatively intact cognitive function (SLUMS 28) and was able to give a
detailed history of her psychiatric illness. She reported that the current medication
regimen helped her more than all other medications she had tried since her 20s, and
she had avoided hospitalization for the last seven years. Ms. S denied any history
suggestive of hypomania or mania and reported a lifelong history of excessive anxiety
and added, “I am a worrier.” The psychiatrist also noted that Ms. S had a history of
recurrent UTIs and was put on antibiotics two days before consultation for another UTI.
On a mental status exam, Ms. S was found to be anxious and eager to please. She
denied any suicidal or homicidal ideas or any current auditory or visual hallucinations.
She acknowledged that she had called her roommate “the devil” but reported that she was
angry with her because she would keep her TV on until late in the night and Ms. S liked
to go to sleep by 8 pm. Staff acknowledged to the psychiatrist that this was an ongoing
conflict between Ms. S and her roommate. Ms. S denied feeling that the staff were taking
her belongings but felt that certain staff were “not nice” to her. The psychiatrist made a
provisional diagnosis of schizoaffective disorder–depressed type and generalized anxiety
disorder and felt that Ms. S’s psychotic symptoms were controlled and hence did not
recommend any change in antipsychotic dosage. Trazodone 50 mg q6 hours for anxiety
was added as needed. The psychiatrist also recommended that the nursing home social
worker address the conflicts between Ms. S and her roommate and requested records
from previous psychiatrists. The social worker, over three meetings with Ms. S and her
roommate, brokered an agreement that Ms. S’s roommate could watch TV in their room
until 9 pm and then go to a family room that had a TV if she wanted to
continue watching.
After four weeks, staff reported that Ms. S’s agitation was much better. She also
reported feeling less anxious and irritable. Ms. S was given as-needed trazodone once
almost daily for the first week but after that staff were able to manage her anxiety without
126
“I Am a Worrier” 127
Teaching Points
Schizoaffective disorder is a psychiatric disorder in which patients demonstrate a com-
bination of symptoms associated with schizophrenia and a mood disorder such as
bipolar disorder or depression. The symptoms of schizophrenia and mood disorder
occur concomitantly for a substantial portion of the illness’s duration. Schizoaffective
disorder can be classified into two subtypes. Schizoaffective disorder–bipolar type
involves both manic and depressive episodes alongside psychotic symptoms.
Schizoaffective disorder–depressive type is characterized by depressive episodes along-
side psychotic symptoms.
Older adults who have a severe persistent mental illness, such as schizoaffective
disorder, are as likely as those who do not have such illness to become agitated due to
medical issues and/or environmental causes. The healthcare provider should consider
these factors before assuming that exacerbation of the underlying psychiatric illness is
the cause of increased psychiatric symptoms. Suppose a resident has been stable on a
psychiatric drug regimen. In that case, it is advisable to avoid any major change in that
regimen because obtaining a similar good therapeutic response from a different drug
regimen is not predictable or assured. The provider should decide to switch antipsycho-
tics or antidepressants only after a thorough review of the patient’s history and perspec-
tives (and those of the involved family) about changes and a review of previous records.
Although drugs like olanzapine and mirtazapine carry substantial metabolic risk (espe-
cially for people who are obese and have diabetes), for some people (such as Ms. S) the
risk may not outweigh the benefit of stabilization of a serious psychiatric illness.
All antipsychotics have the potential to cause weight gain and increase the risk of
obesity and related hyperlipidemia and diabetes. First-generation antipsychotics are used
less now than in decades past. Examples of first-generation antipsychotics include
haloperidol, fluphenazine, perphenazine, and loxapine. This group of medications is
considered relatively low risk for metabolic side effects [1]. The newer atypical anti-
psychotics are well known to be associated with metabolic syndrome and can be thought
of as being high, medium, and low risk for these complications. Medications at high risk
for metabolic syndrome include clozapine and olanzapine. Medications at medium risk
for causing metabolic syndrome include risperidone, quetiapine, paliperidone,
128 Psychotic Disorders
Table 26.1 Atypical antipsychotics and associated risk of metabolic side effects.
asenapine, and iloperidone. Antipsychotics thought to be low risk for metabolic syn-
drome include cariprazine, lurasidone, lumateperone, ziprasidone, pimavanserin,
aripiprazole, and brexpiprazole [2]. See Table 26.1 for details.
Adults with schizophrenia have three and a half times the mortality risk than the
general population, with cardiovascular diseases the most common cause. Antipsychotic
medications with a high or medium risk of inducing metabolic abnormalities are
associated with almost a three-fold higher risk of major cardiovascular events [1].
Regular monitoring of weight, lipid panel, and screening for diabetes is important for
those on long-term antipsychotic treatment.
Take-Home Points
based studies. Advances in Therapy, 38 (5), Hammoudeh, S., Al Lawati, H., Ghuloum, S.,
2491–2512. www.ncbi.nlm.nih.gov/pmc/ Iram, H., Yehya, A., Becetti, I., &
articles/PMC8107077/ Al-Amin, H. (2020). Risk factors of
Carli, M., Kolachalam, S., Longoni, B., metabolic syndrome among patients
Pintaudi, A., Baldini, M., Aringhieri, S., & receiving antipsychotics: A retrospective
Scarselli, M. (2021). Atypical antipsychotics study. Community Mental Health Journal,
and metabolic syndrome: From molecular 56, 760–770.
mechanisms to clinical differences. Sneller, M. H., De Boer, N., Everaars, S.,
Pharmaceuticals, 14 (3), 238. Schuurmans, M., Guloksuz, S., Cahn, W., &
DeJongh, B. M. (2021). Clinical pearls for the Luykx, J. J. (2021). Clinical, biochemical
monitoring and treatment of antipsychotic and genetic variables associated with
induced metabolic syndrome. Mental metabolic syndrome in patients with
Health Clinician, 11 (6), 311–319. schizophrenia spectrum disorders using
second-generation antipsychotics: a
Friedrich, M. E., Winkler, D., Konstantinidis, systematic review. Frontiers in Psychiatry,
A., Huf, W., Engel, R., Toto, S., Grohmann, 12, 625935.
R., & Kasper, S. (2020). Cardiovascular
adverse reactions during antipsychotic Ventriglio, A., Baldessarini, R. J., Vitrani, G.,
treatment: Results of AMSP, a drug Bonfitto, I., Cecere, A. C., Rinaldi, A., &
surveillance program between 1993 and Bellomo, A. (2019). Metabolic syndrome in
2013. The International Journal of psychotic disorder patients treated with oral
Neuropsychopharmacology, 23 (2), 67–75. and long-acting injected antipsychotics.
https://doi.org/10.1093/ijnp/pyz046 Frontiers in Psychiatry, 9, 744.
Section 3 Psychotic Disorders
27 Medication-Induced Delirium
Mr. M, an 82-year-old widowed male who had MNCD due to Alzheimer’s disease, was
transferred from the hospital to a nursing home for rehabilitation after being treated for
syncope due to severe bradycardia with pacemaker implantation and initiation of 200 mg/
day of amiodarone. He started developing changes in mental status a week after discharge,
including worsening disorientation, agitation, insomnia, aggressive behavior during per-
sonal care, and paranoia. Staff at the nursing home requested a psychiatric consult to
facilitate hospitalization to an inpatient psychiatric unit, as Mr. M was “violent.” The
psychiatrist found Mr. M to be agitated and repeatedly yelling, “Get out of my house!”
After an emergency assessment, the consulting psychiatrist diagnosed amiodarone-induced
delirium. Amiodarone was discontinued (after consultation with the resident’s cardiologist)
and Mr. M was given risperidone (0.25 mg twice a day, increased the next day to 0.5 mg twice
a day) for agitation and aggression. The aggression improved dramatically after two days
and resolved completely after seven days. Risperidone was discontinued soon after.
Teaching Points
Delirium is identified when there is an abrupt onset of a condition affecting focus and
consciousness, occurring rapidly and differing from the usual state. This condition tends
to vary in intensity and is accompanied by cognitive deficits (like memory loss, confu-
sion, language issues, visuospatial problems, or perception abnormalities). Delirium can
present with hyperactivity, restlessness, and agitation but can also present as a more
hypoactive variant [1]. Those with hypoactive delirium may be more at risk of delayed
workup as they have fewer easily-recognized symptoms such as sleepiness and social
withdrawal and this variant may be more serious [2] (Figure 27.1).
Older adults are at higher risk of drug-induced (or any other) delirium due to
heightened sensitivity to medications, more comorbidities, and less cognitive reserve.
Older adults with cognitive impairment have an even higher incidence of delirium and
are also more likely to suffer from sensory impairment due to poor vision and hearing.
This can increase the risk of drug-induced delirium. They are more likely to be subject to
polypharmacy, a well-known risk factor for drug-related mental status changes.
Although drugs can be instigators of delirium, a multifactorial model of delirium is
probably more accurate. In this model, even a relatively benign-appearing medication
such as a commonly prescribed antibiotic can lead to delirium in those with a baseline
high vulnerability to the condition.
Amiodarone (and many other prescription drugs) has the potential to cause signifi-
cant adverse cognitive effects in older adults, especially those who have MNCD. Several
130
“Get out of My House” 131
case reports exist in the literature. The unique pharmacokinetics of amiodarone can also
lead to delayed neuropsychiatric symptoms and prolonged symptoms after discontinu-
ation of the medication [3,4].
Medications well known to cause delirium in those with MNCD include
anticholinergics, benzodiazepines, antihistamine medications, steroids, and opiate
analgesics. Less commonly known to cause acute mental status changes are antiparkin-
sonian agents, antibiotics, antihypertensives (b blockers), NSAIDs, anticonvulsants, and
diuretics (Box 27.1).
A review of medications is warranted with each case of delirium in older adults.
A thorough medication review should focus on the considerations detailed in Box 27.2.
Laboratory evaluation of suspected drug-induced delirium can be helpful and is
individualized to the suspected offending agent. Labs potentially important in the
evaluation of suspected drug-induced delirium are shown in Box 27.3.
132 Psychotic Disorders
Box 27.3 Labs that may be helpful in the workup of suspected drug-induced delirium
1. Serum electrolytes. Useful when the patient is on diuretics, lithium, or trimethoprim/
sulfa and there is suspicion of drug-induced electrolyte disturbances.
2. Serum liver function tests. Use especially in cases of suspected medication-induced
hepatic encephalopathy (acetaminophen, amiodarone).
3. Renal function panel. Acute changes in renal function can lead to a buildup of
previously tolerated doses of medication known to cause neuropsychiatric side effects.
4. Thyroid function tests. Hypo or hyperthyroid states triggered by medications such as
lithium and amiodarone can lead to mental status changes.
5. Blood glucose levels. Medications like beta blockers can mask the other physical signs
of hypoglycemia that would otherwise accompany acute mental status changes.
Fluoroquinolone antibiotics are known to increase the risk of hypoglycemia, especially
in older adults.
6. Therapeutic drug levels. Older adults, those with impaired kidney function, and those
with low body weight are especially vulnerable to digoxin toxicity and lithium toxicity.
7. Vitamin levels. Certain drugs (anticonvulsants, methotrexate, diuretics, and antacids)
are well known to influence thiamine, b12, and folate absorption and metabolism.
“Get out of My House” 133
Take-Home Points
28 Hoarding Disorder
Ms. T was a 77-year-old female who had recently moved into Shady Oaks, a local long-
term care facility. Before her move to the facility, she had been living in a trailer home
near the outskirts of her small town. She had never married and had no children. Her
only living relative was a sister who lived several states away and who had limited contact
with Ms. T.
Ms. T had a fall at home about three months before moving to Shady Oaks. She was
unable to reach her phone and was only rescued after a concerned neighbor noted
Ms. T had not been seen feeding the neighborhood cats for a couple of days, which
was her habit. This neighbor was unable to reach Ms. T by phone or get her to answer
her door. Local law enforcement were able to see Ms. T on her bathroom floor through
the window. Paramedics and firefighters were alerted and successful in gaining entry to
the trailer but were nearly unable to reach her due to obstructing debris stacked floor
to ceiling.
It took over an hour to clear a path allowing Ms. T to be extracted from her home.
She was brought to the hospital where she was diagnosed with a hip fracture, rhabdo-
myolysis, and renal failure from her prolonged time on the ground after her fall. After
surgery for her fracture and a seven-day hospital stay, she was transferred to a unit of the
local skilled nursing facility. She recovered somewhat after nearly two weeks of daily
therapy and was able to walk with a walker. Her discharge was complicated because her
home had been condemned by the local building inspector as being unsafe for habita-
tion. The social worker at the facility accompanied the building inspector for a brief visit
to assess the living situation of Ms. T’s prior residence and was shocked to find her home
at least 90% unusable and crammed full of what could only be described as random
discarded items and trash. Ms. T’s living area consisted of a small trail extending from a
dilapidated recliner to both the bathroom and an extremely cluttered and unhygienic
kitchen. Cat feces were found throughout the home and an infestation of roaches
was evident.
Carol, the facility social worker, scheduled a meeting with Ms. T to inform her that
returning to her home was not possible as it had been condemned by the city as
uninhabitable. Ms. T sobbed, “Oh no! What is going to happen to all my things?
I have to have my things! My whole life is in there!” Carol assisted Ms. T with finding
a local contractor to clean the rented trailer but it was more than she could afford.
Arrangements were made for her to stay at Shady Oaks in the long-term care wing of the
facility in a Medicaid-funded room with one roommate.
Her transition to her new home was problematic. Ms. T was extremely private and
refused to let staff into her side of the room, even to do basic cleaning. Her roommate
135
136 Anxiety and Related Disorders
was unhappy as the room became cluttered with various items collected from throughout
the facility that Ms. T thought might later “come in handy for something”. She had
several meetings with the facility administrator about the situation and was forced to
keep her room to an acceptable level of clutter. The facility psychiatrist was asked to
evaluate Ms. T for her “behaviors”.
The interview with Ms. T was initially difficult. She was reserved and somewhat aloof
at first after understanding that the provider was a mental health professional. Her
answers to questions were generally short and direct. She started sobbing when asked
about her life before coming to the care facility. “I miss my cats. I miss my house. I miss
my things. I don’t know why I am here. They had no right!”
She expressed sadness and regret over the loss of her home but seemed to have little
insight into how it came to be condemned as an unsafe dwelling. “I hate being here. This
isn’t living. Everything I own is now in this tiny room. I have nothing!” The examiner
noted that the patient’s small room was already becoming cluttered with items such as
bags of empty food containers, stacks of newspapers, and even sacks of empty toilet
paper rolls.
She expressed that she was very sad and had little-to-no enjoyment in the day-to-day
activities or social interactions at Shady Oaks. She was sleeping poorly, often watching
home shopping networks or infomercials into the early hours of the morning and then
sleeping through breakfast and sometimes through lunch. She felt tired. When asked about
feelings of guilt she admitted that she could not stop thinking about how she had let her life
come to this and felt very sad and guilty for all of the neighborhood cats that she had left
behind. Her appetite was also poor. When asked about suicidal thoughts she said, “I would
never do that, but living like this is pointless.” She admitted to the depression symptoms
starting soon after finding out she could not return to her home three weeks prior.
Ms. T seemed to have at least average intelligence. She scored a 29/30 on her SLUMS
exam and her patient health questionnaire (PHQ-9) score was 22. Her GAD-7 score was 15.
Ms. T was diagnosed with MDD with comorbid hoarding disorder. She was not open
to discussing her tendency to “keep things” but was open to the examiner’s suggestion
that she had clinical depression. She was agreeable to treatment with an antidepressant
and was started on sertraline 25 mg daily, which was increased to 50 mg daily in one
week. She agreed to speak with the facility psychotherapist for therapy once per week.
At a follow-up appointment four weeks later Ms. T seemed a little less depressed. The
nursing staff informed the examiner that she seemed to be adjusting a bit better to the
facility routine and could be seen eating with one or two other women at the facility that
she seemed to have befriended. She was coming out to the dining room for some of her
meals. She told her doctor that she was feeling a little bit better although she would still
cry at least daily when she thought of her home and the things that she had lost. She had
begrudgingly agreed to limit her collected items to one corner of her room and had
started supportive psychotherapy with the on-site licensed clinical social worker. She told
the physician that she liked the visits with the therapist and that it was “nice to have
someone to talk to about everything.”
Teaching Points
Hoarding disorder is now considered one of the obsessive–compulsive (OCD) and
related disorders. It is characterized by the urge to save and acquire new items and the
“My Whole Life Is in There!” 137
difficulty in discarding collected items regardless of value. These urges lead to the
progressive accumulation and clutter of living spaces severe enough to lead to conse-
quences such as unsafe living conditions and the development of impairment in social or
occupational arenas. The condition is often associated with poor insight or even delu-
sional thinking such as beliefs that worthless items will become indispensable in
the future.
The prevalence of hoarding disorder is thought to be about 6% in patients over the
age of 70. The symptoms usually begin in young adulthood and increase in severity with
age. Seniors with hoarding disorder are at risk of multiple adverse outcomes that are
thought to be secondary to the high concurrence of malnutrition, medication misman-
agement, falls, food contamination, and eviction from their homes/housing insecurity
[1]. Approximately two-thirds of patients with hoarding disorder have comorbid psy-
chiatric diagnoses, most commonly mood or anxiety disorders.
Hoarding disorder is considered to be a chronic and progressive disease and most
adults will recall that their hoarding behaviors began before the age of 20.
Epidemiological studies seem to show no real differences in the prevalence of the
disorder between males and females [2].
Those who suffer from hoarding disorder often form strong emotional attachments
to their possessions, making it very difficult for them to throw anything away, even
if items are useless or unsanitary. The behaviors can be distressing and difficult to
control. The disorder can lead to significant impairment in social, occupational, and
family functioning. Patients with this condition may have little-to-no insight into
their condition and others may fully realize the magnitude of their pathology. The
diagnostic features of hoarding disorder as specified in DSM–5–TR are listed in
Box 28.1 [3].
Diagnosing hoarding disorder can be difficult as patients often present late in the
stage of their disease, if at all. Typically, poor to absent insight contributes to late
presentations. These individuals also experience significant shame and societal stigma-
tization related to the condition [4].
The causes of hoarding disorder are not fully known. As with many psychiatric
conditions, genetic predisposition, environmental/developmental factors, and psycho-
logical factors are thought to be at play. Half of those with hoarding disorder will identify
a family member who also hoards [5].
Older adults with hoarding disorder commonly suffer from self-neglect and are at
risk of food contamination, malnutrition, medication mismanagement, falls, and evic-
tion from their homes. In older adults, 25% with hoarding disorder admit to having an
insect infestation in their homes and 58% report that their home is a fire hazard [6].
Comorbid neuropsychiatric illnesses are common, including mood and anxiety dis-
orders, and attention deficit disorder [7].
Treatment options are fairly limited due in part to the fact that the condition has only
been officially recognized as a psychiatric condition with clear diagnostic criteria since
2013. The most effective treatment for older adults with hoarding disorder is cognitive
rehabilitation and exposure/sorting therapy (CREST). This therapy pairs cognitive
training to improve executive functioning with behavioral exposure to the distress of
discarding or not acquiring objects [8]. This therapy is challenging for several reasons,
including its time-consuming nature, limited availability of trained therapists, and the
tendency of some patients to have limited insight into their condition.
138 Anxiety and Related Disorders
Specify if:
With excessive acquisition: If difficulty discarding possessions is accompanied by
excessive acquisition of items that are not needed or for which there is no
available space.
Specify if:
With good or fair insight: The individual recognizes that hoarding-related beliefs and
behaviors (pertaining to difficulty discarding items, clutter, or excessive acquisition)
are problematic.
With poor insight: The individual is mostly convinced that hoarding-related beliefs and
behaviors (pertaining to difficulty discarding items, clutter, or excessive acquisition) are
not problematic despite evidence to the contrary.
With absent insight/delusional beliefs: The individual is completely convinced that
hoarding-related beliefs and behaviors (pertaining to difficulty discarding items, clutter,
or excessive acquisition) are not problematic despite evidence to the contrary.
Take-Home Points
Hoarding disorder is now considered one of the obsessive–compulsive and related disorders. It is
thought to affect about 6% of those over the age of 70.
Symptoms of hoarding disorder are thought to begin in young adulthood and increase in severity
with age.
Those with hoarding disorder are likely to be diagnosed late in the course of their disease due to a
prominent lack of insight, shame, and social stigma.
Complications of hoarding disorder include food contamination, malnutrition, medication misman-
agement, falls, and eviction from the home.
The best treatment outcomes have been shown with CREST. This treatment can be limited by the
availability of appropriately-trained professionals and the lack of insight by patients.
“My Whole Life Is in There!” 139
28 Hoarding Disorder
Ms. T was a 77-year-old female who had recently moved into Shady Oaks, a local long-
term care facility. Before her move to the facility, she had been living in a trailer home
near the outskirts of her small town. She had never married and had no children. Her
only living relative was a sister who lived several states away and who had limited contact
with Ms. T.
Ms. T had a fall at home about three months before moving to Shady Oaks. She was
unable to reach her phone and was only rescued after a concerned neighbor noted
Ms. T had not been seen feeding the neighborhood cats for a couple of days, which
was her habit. This neighbor was unable to reach Ms. T by phone or get her to answer
her door. Local law enforcement were able to see Ms. T on her bathroom floor through
the window. Paramedics and firefighters were alerted and successful in gaining entry to
the trailer but were nearly unable to reach her due to obstructing debris stacked floor
to ceiling.
It took over an hour to clear a path allowing Ms. T to be extracted from her home.
She was brought to the hospital where she was diagnosed with a hip fracture, rhabdo-
myolysis, and renal failure from her prolonged time on the ground after her fall. After
surgery for her fracture and a seven-day hospital stay, she was transferred to a unit of the
local skilled nursing facility. She recovered somewhat after nearly two weeks of daily
therapy and was able to walk with a walker. Her discharge was complicated because her
home had been condemned by the local building inspector as being unsafe for habita-
tion. The social worker at the facility accompanied the building inspector for a brief visit
to assess the living situation of Ms. T’s prior residence and was shocked to find her home
at least 90% unusable and crammed full of what could only be described as random
discarded items and trash. Ms. T’s living area consisted of a small trail extending from a
dilapidated recliner to both the bathroom and an extremely cluttered and unhygienic
kitchen. Cat feces were found throughout the home and an infestation of roaches
was evident.
Carol, the facility social worker, scheduled a meeting with Ms. T to inform her that
returning to her home was not possible as it had been condemned by the city as
uninhabitable. Ms. T sobbed, “Oh no! What is going to happen to all my things?
I have to have my things! My whole life is in there!” Carol assisted Ms. T with finding
a local contractor to clean the rented trailer but it was more than she could afford.
Arrangements were made for her to stay at Shady Oaks in the long-term care wing of the
facility in a Medicaid-funded room with one roommate.
Her transition to her new home was problematic. Ms. T was extremely private and
refused to let staff into her side of the room, even to do basic cleaning. Her roommate
135
136 Anxiety and Related Disorders
was unhappy as the room became cluttered with various items collected from throughout
the facility that Ms. T thought might later “come in handy for something”. She had
several meetings with the facility administrator about the situation and was forced to
keep her room to an acceptable level of clutter. The facility psychiatrist was asked to
evaluate Ms. T for her “behaviors”.
The interview with Ms. T was initially difficult. She was reserved and somewhat aloof
at first after understanding that the provider was a mental health professional. Her
answers to questions were generally short and direct. She started sobbing when asked
about her life before coming to the care facility. “I miss my cats. I miss my house. I miss
my things. I don’t know why I am here. They had no right!”
She expressed sadness and regret over the loss of her home but seemed to have little
insight into how it came to be condemned as an unsafe dwelling. “I hate being here. This
isn’t living. Everything I own is now in this tiny room. I have nothing!” The examiner
noted that the patient’s small room was already becoming cluttered with items such as
bags of empty food containers, stacks of newspapers, and even sacks of empty toilet
paper rolls.
She expressed that she was very sad and had little-to-no enjoyment in the day-to-day
activities or social interactions at Shady Oaks. She was sleeping poorly, often watching
home shopping networks or infomercials into the early hours of the morning and then
sleeping through breakfast and sometimes through lunch. She felt tired. When asked about
feelings of guilt she admitted that she could not stop thinking about how she had let her life
come to this and felt very sad and guilty for all of the neighborhood cats that she had left
behind. Her appetite was also poor. When asked about suicidal thoughts she said, “I would
never do that, but living like this is pointless.” She admitted to the depression symptoms
starting soon after finding out she could not return to her home three weeks prior.
Ms. T seemed to have at least average intelligence. She scored a 29/30 on her SLUMS
exam and her patient health questionnaire (PHQ-9) score was 22. Her GAD-7 score was 15.
Ms. T was diagnosed with MDD with comorbid hoarding disorder. She was not open
to discussing her tendency to “keep things” but was open to the examiner’s suggestion
that she had clinical depression. She was agreeable to treatment with an antidepressant
and was started on sertraline 25 mg daily, which was increased to 50 mg daily in one
week. She agreed to speak with the facility psychotherapist for therapy once per week.
At a follow-up appointment four weeks later Ms. T seemed a little less depressed. The
nursing staff informed the examiner that she seemed to be adjusting a bit better to the
facility routine and could be seen eating with one or two other women at the facility that
she seemed to have befriended. She was coming out to the dining room for some of her
meals. She told her doctor that she was feeling a little bit better although she would still
cry at least daily when she thought of her home and the things that she had lost. She had
begrudgingly agreed to limit her collected items to one corner of her room and had
started supportive psychotherapy with the on-site licensed clinical social worker. She told
the physician that she liked the visits with the therapist and that it was “nice to have
someone to talk to about everything.”
Teaching Points
Hoarding disorder is now considered one of the obsessive–compulsive (OCD) and
related disorders. It is characterized by the urge to save and acquire new items and the
“My Whole Life Is in There!” 137
difficulty in discarding collected items regardless of value. These urges lead to the
progressive accumulation and clutter of living spaces severe enough to lead to conse-
quences such as unsafe living conditions and the development of impairment in social or
occupational arenas. The condition is often associated with poor insight or even delu-
sional thinking such as beliefs that worthless items will become indispensable in
the future.
The prevalence of hoarding disorder is thought to be about 6% in patients over the
age of 70. The symptoms usually begin in young adulthood and increase in severity with
age. Seniors with hoarding disorder are at risk of multiple adverse outcomes that are
thought to be secondary to the high concurrence of malnutrition, medication misman-
agement, falls, food contamination, and eviction from their homes/housing insecurity
[1]. Approximately two-thirds of patients with hoarding disorder have comorbid psy-
chiatric diagnoses, most commonly mood or anxiety disorders.
Hoarding disorder is considered to be a chronic and progressive disease and most
adults will recall that their hoarding behaviors began before the age of 20.
Epidemiological studies seem to show no real differences in the prevalence of the
disorder between males and females [2].
Those who suffer from hoarding disorder often form strong emotional attachments
to their possessions, making it very difficult for them to throw anything away, even
if items are useless or unsanitary. The behaviors can be distressing and difficult to
control. The disorder can lead to significant impairment in social, occupational, and
family functioning. Patients with this condition may have little-to-no insight into
their condition and others may fully realize the magnitude of their pathology. The
diagnostic features of hoarding disorder as specified in DSM–5–TR are listed in
Box 28.1 [3].
Diagnosing hoarding disorder can be difficult as patients often present late in the
stage of their disease, if at all. Typically, poor to absent insight contributes to late
presentations. These individuals also experience significant shame and societal stigma-
tization related to the condition [4].
The causes of hoarding disorder are not fully known. As with many psychiatric
conditions, genetic predisposition, environmental/developmental factors, and psycho-
logical factors are thought to be at play. Half of those with hoarding disorder will identify
a family member who also hoards [5].
Older adults with hoarding disorder commonly suffer from self-neglect and are at
risk of food contamination, malnutrition, medication mismanagement, falls, and evic-
tion from their homes. In older adults, 25% with hoarding disorder admit to having an
insect infestation in their homes and 58% report that their home is a fire hazard [6].
Comorbid neuropsychiatric illnesses are common, including mood and anxiety dis-
orders, and attention deficit disorder [7].
Treatment options are fairly limited due in part to the fact that the condition has only
been officially recognized as a psychiatric condition with clear diagnostic criteria since
2013. The most effective treatment for older adults with hoarding disorder is cognitive
rehabilitation and exposure/sorting therapy (CREST). This therapy pairs cognitive
training to improve executive functioning with behavioral exposure to the distress of
discarding or not acquiring objects [8]. This therapy is challenging for several reasons,
including its time-consuming nature, limited availability of trained therapists, and the
tendency of some patients to have limited insight into their condition.
138 Anxiety and Related Disorders
Specify if:
With excessive acquisition: If difficulty discarding possessions is accompanied by
excessive acquisition of items that are not needed or for which there is no
available space.
Specify if:
With good or fair insight: The individual recognizes that hoarding-related beliefs and
behaviors (pertaining to difficulty discarding items, clutter, or excessive acquisition)
are problematic.
With poor insight: The individual is mostly convinced that hoarding-related beliefs and
behaviors (pertaining to difficulty discarding items, clutter, or excessive acquisition) are
not problematic despite evidence to the contrary.
With absent insight/delusional beliefs: The individual is completely convinced that
hoarding-related beliefs and behaviors (pertaining to difficulty discarding items, clutter,
or excessive acquisition) are not problematic despite evidence to the contrary.
Take-Home Points
Hoarding disorder is now considered one of the obsessive–compulsive and related disorders. It is
thought to affect about 6% of those over the age of 70.
Symptoms of hoarding disorder are thought to begin in young adulthood and increase in severity
with age.
Those with hoarding disorder are likely to be diagnosed late in the course of their disease due to a
prominent lack of insight, shame, and social stigma.
Complications of hoarding disorder include food contamination, malnutrition, medication misman-
agement, falls, and eviction from the home.
The best treatment outcomes have been shown with CREST. This treatment can be limited by the
availability of appropriately-trained professionals and the lack of insight by patients.
“My Whole Life Is in There!” 139
Mrs. U was a 96-year-old widowed woman who was admitted to the nursing home
because of frailty, blindness due to macular degeneration, and recurrent falls. She had a
sister who was two years younger and in good health, one son, three grandchildren, eight
great-grandchildren, and one great-great-grandchild. For eight to nine months
Mrs. U had been having increased anxiety and nervousness, had used her call light
excessively, was shouting “help, help” for long periods of time, and was grabbing passers-
by and asking them to help her.
Mrs. U had been in the long-term care facility for several years and had always been
somewhat anxious, but these symptoms were much more severe and triggered by a
severe UTI nine months earlier. The staff was initially able to manage her behavior with
psychosocial approaches, such as taking her to church service twice a day (Mrs. U was a
Methodist and religious but did not mind attending church services of different denom-
inations), hand massage, and soothing music. However, over five months, the symptoms
became severe and difficult to manage. Mrs. U usually slept well and, although she had
been eating less for a few weeks, had no weight loss.
The family started decreasing their visits because they felt Mrs. U became more agitated
when they visited and asked her how she was feeling. The family felt helpless when they
could not calm her down and the staff told them that she was having “another bad day.”
The primary care physician (PCP) had tried citalopram and mirtazapine with her,
but Mrs. U could not tolerate either of these medications. Hence, the PCP referred her to
the consulting psychiatrist. The psychiatrist found Mrs. U to be pleasant and talkative,
and she anxiously stated, “My vision has been worsening in the last two weeks.” She
described herself as being a “worrier,” and her son confirmed that she had always been
extremely nervous and impatient, would be “easily stressed,” and thought and feared the
worst in any situation. Mrs. U denied any depressive symptoms, and her son confirmed
that she had not had any significant depressive symptoms in the past.
Mrs. U’s PCP evaluated her for a UTI, constipation, pain, electrolyte imbalance,
thyroid dysfunction, and vitamin deficiency and did not find any problems. Mrs. U’s
vision complaints were not new and were due to macular degeneration, which was being
addressed by the ophthalmologist. The pharmacist reviewed Mrs. U’s medications and
recommended discontinuing cyclobenzaprine (which the PCP subsequently did) but did
not find any other medication that could cause anxiety. Mrs. U’s SLUMS score was 24
(indicating relatively intact cognitive function, given her age and vision problems), and
her Geriatric Depression Scale (GDS) score was 5 (suggesting minimal depression). The
psychiatrist diagnosed Mrs. U as having general anxiety disorder (GAD) and mild
140
“My Vision Has Been Worsening in the Last Two Weeks” 141
Teaching Points
It is not uncommon for a resident to try multiple medications to manage anxiety
disorder before finding a tolerated and effective psychotropic. Although an SSRI or
SNRI is usually the first-line agent for pharmacotherapy of GAD, the healthcare profes-
sional should also consider prescribing buspirone. The full effects of medication for
anxiety disorder may take a few months. Specific guidance and counseling of family and
staff regarding psychosocial approaches have a much higher success rate than
nonspecific recommendations.
Generalized anxiety disorder is the most commonly encountered anxiety disorder.
Other anxiety disorders include panic disorder, agoraphobia, social anxiety disorder, and
specific phobias. Symptoms of GAD include chronic and pervasive anxiety and excessive
worry. These can be accompanied by various physical symptoms such as increased heart
rate, shortness of breath, muscle tension, sweating, GI symptoms, dizziness, sleep
disturbances, fatigue, and changes in appetite.
Older adults diagnosed with GAD often have a lifelong history of excessive worry and
anxiety, which will usually be corroborated by family members or caregivers. Diagnosing
the condition is relatively straightforward. Older adults able to participate in a history
may describe feelings of anxiety, worry, irritability, difficulty sleeping, and a variety of
physical symptoms that are difficult to attribute to other specific medical conditions.
Concern for anxiety is often brought up by caregivers when patients demonstrate
disruptive behaviors related to anxiety, such as yelling out, resisting care, becoming
frightened with personal care, and repetitive behaviors such as pushing a call light many
times a day, pacing, or restlessness.
Those with GAD may have frequent exacerbations of their condition that can be
triggered by changes in routine, loneliness, boredom, new staff, changes in medical
conditions, and pain. Those older adults with more severe GAD may be triggered by
the smallest inconveniences or irritants.
Diagnosing GAD is possible with a good history, mental status exam, review of
medications, and review of pertinent labs. Cognitively intact patients will often readily
and eagerly describe their anxiety and may describe an inability to enjoy day-to-day
activities due to excessive worry. Perseveration may be prominent. The family may tell
142 Anxiety and Related Disorders
providers that their loved one will call excessively or be excessively focused on physical
symptoms and their potentially dire implications. The mental status exam is usually
positive for an anxious mood. The workup should include pertinent labs such as thyroid
status, electrolyte status, b12, and vitamin D levels. A brief neurological exam focusing
on the presence or absence of focal neurologic signs, tremors, and gait is helpful.
Recognizing and diagnosing anxiety in an older adult with advanced MNCD can be
more challenging. These patients often cannot express themselves sufficiently to tell the
clinician or caregiver their feelings. The provider can then look for outward signs of
anxiety or ask for detailed behavior descriptions from caregivers. Those with advanced
MNCD may show signs of restlessness, which can be seen as pacing, wringing of hands,
or fidgeting. Anxious behaviors can also be seen to include frequent nonsensical verbal-
izations or vocalizations. Patients who are anxious are often easily irritated, may lash out
verbally or physically at times, and may have a hard time getting to sleep at night. Some
other signs of anxious behavior in those with severe cognitive impairment are shown in
Box 29.1.
The differential diagnosis for general and persistent anxiety symptoms can include
endocrine causes such as overreplacement of thyroid hormone and functioning thyroid
nodules. Very rarely catecholamine-producing neuroendocrine tumors can lead to
anxiety symptoms, tachycardia, and episodic hypertension. Symptoms of hyper or
hypoglycemia can be mistaken for anxiety. Cardiac causes such as uncontrolled atrial
fibrillation can lead to tachycardia and shortness of breath that may be mistaken for
anxiety symptoms. Those with chronic pulmonary disease often deal with anxiety
secondary to chronic shortness of breath, hypoxia, and hypercapnia.
Once the diagnosis of GAD is made treatment options can be considered. First-line
agents for the treatment of GAD are SSRIs. Of the six SSRIs commonly prescribed
sertraline and fluoxetine may be preferred due to lack of dosing restrictions from a
cardiac standpoint and lack of anticholinergic effects. Serotonin–norepinephrine reup-
take inhibitors are also a good choice and include duloxetine and venlafaxine. In those
who may have a contraindication or intolerance to SSRIs or SNRIs, there are several
good alternatives. Buspirone is generally well tolerated in older adults and doses should
quickly be titrated up to 30 or 60 mg daily. It may then take weeks to have a full clinical
response. Mirtazapine is a similarly well-tolerated second-line medication. A summary
of some commonly used pharmaceuticals used in anxiety is listed in Box 29.2.
“My Vision Has Been Worsening in the Last Two Weeks” 143
Second-line agents
1. Buspirone. The dose should be titrated to 30 or 60 mg daily. May take weeks for
clinical response.
2. Mirtazapine. May take weeks for clinical response. Good if increased appetite is desired.
3. Propranolol. Even low doses of propranolol are unlikely to have a cardiovascular effect
and can be therapeutic for anxiety. Doses for anxiety are generally given at
10–20 mg bid.
4. Trazodone. Mildly sedating antidepressants in low doses can be helpful for anxiety.
Relatively quick acting. May be a good bridge medication when waiting for SSRI or
SNRI therapeutic effect.
Avoid
1. Benzodiazepines. Helpful in the short-term in younger patients but should be avoided
in older adults.
2. Anticholinergics. Drugs like hydroxyzine are helpful in younger adults but should be
avoided in older adults.
3. Antipsychotics. Although these drugs can have a calming effect, they should be
avoided in the absence of psychosis due to unfavorable side effect profiles and black
box warnings regarding use in older adults.
Box 29.4 Considerations for those that do not adequately respond to first-line
treatment for GAD
1. Consider the dose of the first-line agent. Often the target dose for anxiety disorders is
higher than that needed for response in depression and related disorders. Some
providers are overly cautious about titrating the dose of first-line agents to a truly
therapeutic dose.
2. Consider adding an augmenting agent to the SSRI or SNRI. For example, add buspirone
or trazodone to sertraline or fluoxetine.
3. Switch from an SSRI to an SNRI. Some patients respond better to the mechanism of an
SNRI than SSRI.
4. Consider the possibility of psychotic symptoms, especially in those with MNCDs. Soft
signs of psychosis in older adults may be an extreme fear response to relatively benign
stimuli, talking to oneself (responding to inner stimuli), picking at one’s skin as if bugs
or irritants were present, or the presence of very disorganized or bizarre behavior.
It may be that SSRIs or SNRIs are not adequate for these patients. A trial of a low dose of
atypical antipsychotic may be helpful.
Follow up for those initiating treatment for GAD should be relatively frequent until
symptoms are stabilized. Resist the urge to change first-line treatments before an
adequately long trial (eight weeks) at an adequate dose has been accomplished.
Attempts should be made to taper antipsychotics at least quarterly in those patients that
may have benefited from their use for anxiety contributed to by psychosis in MNCD.
Take-Home Points
It is not uncommon for a resident to try multiple medications to manage an anxiety disorder before
finding a tolerated and effective psychotropic. An SSRI or SNRI is usually the first-line agent for
pharmacotherapy and GAD.
Second-line agents or supporting agents may include buspirone, mirtazapine, propranolol, and
trazodone. Avoid benzodiazepines and anticholinergics.
Concern for anxiety is often brought up by caregivers when patients demonstrate disruptive
behaviors related to anxiety such as yelling out, resisting care, becoming frightened with personal
care, and repetitive behaviors such as pushing a call light many times a day, pacing, or restlessness.
Strategies to address difficult-to-manage anxiety include reconsidering the first-line agent for
appropriate dose and duration, adding an augmenting agent, switching from an SSRI to an SNRI,
and considering the possibility of low-grade psychosis as a potential contributing factor to anxiety.
“My Vision Has Been Worsening in the Last Two Weeks” 145
30 Panic Disorder
Ms. F was a 70-year-old resident who had a long history of panic disorder with
agoraphobia. She had been stable for decades, taking 5 mg of diazepam twice daily
for the last 30 years, along with 0.25 mg of alprazolam as needed once a day and
imipramine 25 mg daily at bedtime. She was admitted to a nursing facility for rehabili-
tation after receiving a second kidney transplant. Her PCP, who had prescribed her
psychiatric medications for decades, had recently passed away and a young family
practitioner took over her care. He felt that Ms. F carried a high risk of falls and
confusion due to diazepam, as-needed alprazolam, and imipramine. He also felt that
Ms. F had become addicted to diazepam and scheduled a tapering schedule for it.
Within a few weeks, Ms. F was severely agitated and required hospitalization in a
psychiatric unit. She clearly told the psychiatrist, “I am not addicted to Valium.” The
psychiatrist discussed treatment options with Ms. F, including switching to an SSRI,
mirtazapine, or nortriptyline versus reinstating her original medications. Ms. F chose
to restart her original medications, as they had helped her for many years and she was
willing to risk the potential adverse effects, including cardiac risks, falls, and cognitive
impairment. The psychiatrist reinstated Ms. F’s original medication regimen and she
was back to her baseline level of functioning within a couple of weeks. The psychiatrist
also collaborated with the new PCP and explained the severity of the panic disorder
and the plan for frequent outpatient follow-up visits to closely monitor risks for falls
and cognitive impairment. The PCP agreed, as long as the psychiatrist would continue
to manage Ms. F’s panic disorder.
Teaching Points
Panic disorder is a type of anxiety disorder characterized by recurring and unexpected
panic attacks. Panic attacks are periods of intense and overwhelming fear that can be
accompanied by physical symptoms such as rapid heartbeat, chest pain, dizziness,
shortness of breath, tremors, and sweating. The symptoms of these attacks usually
escalate very rapidly and may last anywhere from several minutes to an hour or more.
People with this condition often live in fear of experiencing another panic attack, which
can lead to continuous anxiety and avoidance behaviors. Avoidance behaviors are an
attempt to avoid situations or places in which previous panic attacks have occurred.
These avoidance behaviors can lead to significant disability in the areas of work,
relationships, and general enjoyment of life. Some with panic disorder have comorbid
agoraphobia, which is a fear of being in situations where escape might be difficult
or embarrassing.
146
“I Am Not Addicted to Valium” 147
Box 30.1 DSM–5–TR diagnostic criteria for the diagnosis of panic disorder
1. Recurrent unexpected panic attacks. A panic attack is an abrupt surge of intense fear or
intense discomfort that reaches a peak within minutes and can occur from a calm or an
anxious state. During an attack, four (or more) of the following symptoms occur:
a. Palpitations, pounding heart, or accelerated heart rate.
b. Sweating.
c. Trembling or shaking.
d. Sensations of shortness of breath or smothering.
e. Feelings of choking.
f. Chest pain or discomfort.
g. Nausea or abdominal distress.
h. Feeling dizzy, unsteady, lightheaded, or faint.
i. Chills or heat sensations.
j. Paresthesias (numbness or tingling sensations).
k. Derealization (feelings of unreality) or depersonalization (being detached
from oneself ).
l. Fear of losing control or “going crazy”.
m. Fear of dying.
Note: Culture-specific symptoms (e.g., tinnitus, neck soreness, headache,
uncontrollable screaming or crying) may be seen. Such symptoms should not count as
one of the four required symptoms.
2. At least one of the attacks has been followed by one month (or more) of one or both of
the following:
a. Persistent concern or worry about additional panic attacks or their consequences
(e.g. losing control, having a heart attack, “going crazy”).
b. A significant maladaptive change in behavior related to the attacks (e.g. behaviors
designed to avoid having panic attacks, such as avoidance of exercise or
unfamiliar situations).
effects and the abuse potential of benzodiazepines. Benzodiazepines have been used for
many years for the short-term treatment of panic attacks. Prescribing patterns for
benzodiazepines have changed over the decades as knowledge has increased regarding
the consequences of long-term use. It is not uncommon to encounter older adults who
have been on moderate to high doses of benzodiazepines for many years. These patients
may be extremely fearful of tapering benzodiazepines due to psychological and physio-
logical dependence that often develops. The treatment options for panic disorder are
demonstrated in Box 30.2 [7,8,9].
“I Am Not Addicted to Valium” 149
Box 30.2 Pharmacologic treatment options for those with panic disorder
Acute short-term treatment
Benzodiazepines are recommended for short-term acute treatment of panic disorder.
The effects of benzodiazepines on decreasing the severity and frequency of panic
symptoms can be seen in as little as days to weeks. They are especially helpful as long-
term therapies may take weeks or months for full benefits to be realized. A scheduled
dosing is preferred over as-needed dosing.
Long-term therapies
1. SSRIs: First-line agents. Approved medications safe for use in older adults include
sertraline and fluoxetine. Avoid paroxetine due to anticholinergic effects in older
adults. Doses of citalopram and escitalopram are limited by potential cardiac effects.
It may take weeks for clinical response. May require higher doses than typically seen for
response in depression.
2. SNRIs: Venlafaxine, duloxetine. Effective, but withdrawal syndromes can be
problematic if medications need to be tapered or stopped.
3. Benzodiazepines such as lorazepam, clonazepam. May be required for short-term relief
of panic symptoms and for those with inadequate response to SSRIs and SNRIs.
Generally avoided in older adults but may be hard to stop in those who have had good
responses for years. Use the lowest effective dose [7].
Follow-up for panic disorder should include clinicians asking about the frequency
and severity of panic symptoms, the effectiveness of abortive treatments, and continued
tolerance of long-term medications. It is important to speak with caregivers in older
adults with cognitive impairments.
There are several challenges regarding the treatment of panic disorder that are more
specific to older adults. Benzodiazepine use is a well-accepted abortive treatment for
panic symptoms in younger adults without a history of substance use disorder. They are
fast-acting and generally considered quite safe when used as prescribed. Their use in
older adults is more likely to have unwanted side effects such as increased fall risk,
worsening cognition, and behavioral disinhibition. For these reasons, every effort should
be made to gradually taper older adults off as-needed or scheduled benzodiazepines if
possible. Small reductions in dose are likely to be well tolerated but the final discontinu-
ation of even a tiny dose of benzodiazepine may lead to an acute decompensation in
panic or anxiety symptoms. For this reason, it may be more prudent to taper the dose of
benzodiazepine to the lowest dose possible that does not exacerbate the underlying
anxiety/panic. Older adults may by necessity remain on very low doses of benzodiazep-
ines almost indefinitely. Clinicians should not be so averse to benzodiazepine use in
older adults as to risk psychiatric decompensation and potential hospitalization. Patients
tapering down on benzodiazepines may need up-titration of non-benzodiazepine thera-
peutics like SSRIs, SNRIs, buspirone, trazodone, gabapentin, and so on.
Those with advanced MNCDs are at higher risk of psychotic symptoms that may
trigger anxiety and panic. These patients may benefit from antipsychotic therapy to
address this. The lowest dose possible to control psychotic symptoms should be pre-
scribed and periodic trials of tapering the antipsychotic are important due to increased
mortality risk when used in older adults with MNCDs.
150 Anxiety and Related Disorders
Take-Home Points
Panic disorder is a type of anxiety disorder characterized by recurring and unexpected panic
attacks. Panic attacks are periods of intense and overwhelming fear that can be accompanied by
physical symptoms such as rapid heartbeat, chest pain, dizziness, shortness of breath, tremors,
and sweating.
Benzodiazepines are recommended for short-term acute treatment of panic disorder. The
effects of benzodiazepines on decreasing the severity and frequency of panic symptoms can
be seen in as little as days or weeks. They are especially helpful as long-term therapies may take
weeks or months for full benefits to be realized. A scheduled dosing is preferred over as-
needed dosing.
Pharmacologic treatment options for those with panic disorder can include a short course of
benzodiazepines and long-term SSRIs or SNRIs. Cognitive behavioral therapy has proven to be a
particularly effective nonpharmacologic approach to the treatment of anxiety and panic.
Those with advanced MNCDs are at higher risk of psychotic symptoms that may trigger anxiety and
panic. These patients may benefit from antipsychotic therapy to address this.
Mrs. B was a 91-year-old woman admitted to an AL home six months ago because of
increasing agitation and anxiety. Mr. B, who was 93 years old, could no longer take care
of her. Mrs. B had been diagnosed with MNCD due to Alzheimer’s disease three years
previously. She could not tolerate ChEis due to nausea and diarrhea and had been taking
memantine 10 mg twice daily for two years.
Over the last year, Mrs. B had become increasingly anxious and agitated and would
start yelling for her husband if she was left alone for even a few minutes. Mrs. B would
worry that something terrible had happened to Mr. B and she would not allow him to go
anywhere, even if one of their three daughters agreed to stay with her. Mrs. B would try
to leave the house to look for her husband and would become aggressive if someone tried
to stop her. She showed the same behavior in the AL home, and although lorazepam
0.5 mg three times a day prescribed by her PCP had helped, she became more anxious
when started on sertraline 50 mg daily.
The family was growing increasingly frustrated because they would often spend
hours with Mrs. B without improvement in her anxiety. Mr. B would become very angry
with her, and her daughters could not understand why she was “so stubborn.” A geriatric
psychiatrist was consulted at this point. The psychiatrist learned from Mrs. B’s sister that
they had lost their younger brother through accidental drowning when Mrs. B was ten
years old. Mrs. B was the oldest of six children and did not have any role in the drowning.
Her mother was overwhelmed with working on the farm and raising six children and had
put a lot of responsibility on Mrs. B for housework and looking after her younger
siblings.
Mrs. B had expressed guilt off and on for years over the death and was described by
her family as having been an anxious and shy person who liked to be in her home and
take care of her husband and three daughters. There was no history of her anxiety and
shyness having caused significant impairment in daily functioning in her younger years.
Mrs. B was otherwise in good health and denied any depressive symptoms during the
interview. She reported that she had a difficult life, that her worries about her husband
were “normal,” and added, “Of course, I worry about him.” Mrs. B denied her husband’s
report that she would become “belligerent.” She could not give a detailed history because
of her MNCD, her SLUMS score was 16, and her GDS–15 score was seven. A score over
five is suggestive of depression. The psychiatrist diagnosed severe anxiety disorder and
mild depression in the setting of MNCD. Mrs. B was started on 12.5 mg of sertraline,
which was increased every two weeks to a total of 50 mg daily. The psychiatrist informed
the care staff at the AL home about Mrs. B’s childhood trauma, and both the staff and
family were also counseled that MNCD made Mrs. B more susceptible to emotional
152
“Of Course, I Worry about Him” 153
disorders because of past trauma. The staff became more sympathetic, allowed the
medication more time to work, and tried harder to distract Mrs. B during her times of
heightened anxiety. The family and staff were educated that SSRIs such as sertraline may
cause an initial increase in anxiety before improvements are noted, especially in a patient
who is already anxious.
After eight weeks, the staff and family reported a mild improvement in Ms. B’s
agitation and anxiety, especially her episodes of yelling and disrupting the environment.
The psychiatrist further increased sertraline to 62.5 mg daily for two weeks and then to
75 mg daily. After eight more weeks, the anxiety and agitation were substantially less,
and yelling episodes were occasional and easily managed. The lorazepam was gradually
decreased and changed to as-needed. Mrs. B used it on average once or twice a week.
Teaching Points
Uncontrolled anxiety can adversely affect the ability of an older adult with MNCD to
remain successful in a home setting. Anxiety can be very fatiguing to family members and
other caregivers as well. Older adults with anxiety and MNCD often have a difficult time
with verbal expression, which can make it even more difficult for caregivers to know how
to help with anxiety symptoms. Chronic anxiety is highly associated with depression and
improvements in anxiety often correlate with improvements in depressive symptoms.
The prevalence of mood and anxiety symptoms in those with MNCD is quite high.
A study in 2016 by Zhao et al. showed a prevalence of anxiety in 39% of those with
Alzheimer’s disease [1]. Another study by Chi et al. in 2015 demonstrated a prevalence of
depression of 6–42% depending on the criteria used. It is suggested that depression may
be underestimated in those with severe MNCDs as the underlying disease process can
lead to a declining ability to both have and express depressive or anxious thoughts [2].
A large meta-analysis examining the prevalence of depression, anxiety, and PTSD in the
four most common dementias (Alzheimer’s disease, VaD, DLB, and FTD) showed that
25% of these patients were found to have clinically significant levels of depressive
symptoms and 14% showed clinically significant anxiety [3].
The prevalence of anxiety likely declines as MNCDs progress in the late stages. It is
thought that this may be due to the decline, insight, and awareness that accompanies
these later stages of the disease [4].
The pathophysiology of anxiety involves the dysregulation of several neurotransmit-
ter pathways in the CNS. Neurotransmitters implicated include norepinephrine, sero-
tonin, dopamine, and GABA. This dysregulation leads to dysfunction of the sympathetic
autonomic nervous system as well, which can lead to physical symptoms such as
tachycardia, tachypnea, tremulousness, and elevated blood pressure, for example [5].
The amygdala and limbic system play a pivotal role in controlling anxiety. These
structures are connected to the prefrontal cortex. These neural pathways are targets of
pharmacologic interventions.
Psychosocial factors influence the experience of anxiety in older adults with or
without MNCD. Some factors shown to increase anxiety include very high levels of
social contact, dysfunctional patient–caregiver relationships, and high physical depend-
ency [6]. Other factors that can negatively impact anxiety in older adults include
boredom, social isolation, and unmet physical needs for proper nutrition, warmth, and
cleanliness, for example.
154 Anxiety and Related Disorders
The diagnosis of anxiety in older adults in long-term care should include a physical
exam and basic laboratory testing to detect or rule out physical disorders that can cause,
exacerbate, or mimic anxiety. See Box 31.1 for medical mimickers of anxiety.
A thorough review of medications is important as some medications are known to
influence anxiety. Stimulants are not often used in older adults but are well known to
cause or increase anxiety. Corticosteroids are more often seen in older adults and can
negatively influence anxiety even at relatively low doses. Some antidepressants are known
to cause anxiety, particularly in the initial weeks of treatment. Inhaled and oral medica-
tions for asthma such as albuterol and theophylline are also well known to cause anxiety.
Some patients can become more sensitive to the effects of caffeine as they get older, and
patterns of caffeine use should be explored (Box 31.2).
The treatment of anxiety disorders in older adults in long-term care settings should
consider both environmental and pharmacologic interventions. Those who work regu-
larly with older adults with MNCDs will know that an environment that is familiar and
consistent, calm and quiet, safe, with protected personal space can go a long way to lessen
the symptoms of anxiety. Environmental interventions that may help reduce symptoms
of anxiety are listed in Box 31.3.
The pharmacologic treatment of anxiety in older adults in long-term care shares
similarities with the treatment of anxiety in younger patients. Both SSRIs and SNRIs
are good first-line agents and are generally well tolerated. Selective serotonin reuptake
Box 31.1 Medical conditions that can mimic anxiety in older adults
1. Infections. Urinary tract infections are well known to cause an increase in agitation and
anxiety in older adults.
2. Chronic pain. Undertreated pain can lead to an increase in depression and
anxiety symptoms.
3. Endocrinologic disorders. Hyper or hypoglycemia and hyperthyroid conditions can
mimic anxiety. Pheochromocytoma is a rare cause of anxiety.
4. Cardiovascular conditions such as tachyarrhythmias.
5. Pulmonary conditions associated with dyspnea can trigger anxiety symptoms.
6. Neurological conditions associated with anxiety include but are not limited to seizure
disorder, multiple sclerosis, and TBI.
7. Substance use or withdrawal. The use or tapering of benzodiazepines, opioids, and
alcohol is associated with anxiety.
Box 31.3 Environmental interventions that may help reduce symptoms of anxiety in
older adults in long-term care
1. Familiar and consistent environment. Avoid changes to the environment. Have familiar
objects/furniture in private spaces.
2. Adequate lighting. Well-lit private and public spaces with natural light.
3. Noise control. A calm and quiet living space.
4. Safety. Ensure that the environment has appropriate safety features like handrails and
nonslip flooring, for example.
5. Structured and predictable daily routine.
6. Personalized spaces. Familiar objects and personal items can trigger positive memories
and reduce anxiety.
7 Supportive social environment that includes visits from loved ones, group activities,
and other opportunities for social interaction.
Take-Home Points
Some factors shown to increase anxiety include very high levels of social contact, dysfunctional
patient–caregiver relationships, and high physical dependency. Other factors that can negatively
impact anxiety in older adults include boredom, social isolation, and unmet physical needs for
proper nutrition, warmth, and cleanliness, for example.
As-needed medications for anxiety can be helpful in the initial weeks of treatment as the thera-
peutic effect of SSRIs and SNRIs is approached. Useful medications for as-needed treatment of
anxiety include low-dose trazodone (25 or 50 mg every four to six hours as needed), as well as
gabapentin, mirtazapine, or low-dose propranolol.
Pharmacologic treatment options for those with panic disorder can include a short course of
benzodiazepines and long-term SSRIs or SNRIs. Cognitive behavioral therapy has proven to be a
particularly effective nonpharmacologic approach to the treatment of anxiety and panic.
There is a relatively high comorbidity of psychosis in those with MNCDs, especially in later stages.
Psychosis may be more difficult to detect in those with psychosis in the later stages of neurode-
generative disease as the ability to verbalize experiences becomes more difficult. Anxiety can be
difficult to control in the setting of untreated psychosis.
32 PTSD
Mr. M was a 92-year-old resident living in a long-term care facility. He had been
experiencing nightmares, verbal and physical aggression, anxiety, and hypervigilance.
He would also shout, “Bombs are falling! Run, run!”, thereby agitating other residents,
and had been isolating himself for several weeks. These symptoms started after
Mr. M watched images of recent terrorist attacks on TV. The staff at the nursing home
felt that he should be hospitalized in a psychiatric unit, as he was “psychotic.” The
consulting psychiatrist was asked to help facilitate the hospitalization.
The psychiatrist made an emergency psychiatric evaluation of Mr. M, whose wife
reported that he was an army infantryman from 1942 to 1945. He had been in prolonged,
intense combat in Sicily and Normandy and experienced mild PTSD symptoms and
severe depression from 1946 to 1949. Symptoms gradually decreased after he started
meeting regularly with a group of friends who were also in World War II. Married for 57
years, Mr. M had four children and 11 grandchildren. He had a successful career as a
banker and never abused drugs or alcohol. His wife reported that Mr. M was an
easygoing person who seemed to enjoy life until he entered the nursing home due to
multiple medical problems.
He developed severe peripheral vascular disease, resulting in bilateral above-knee
amputation of his legs for the treatment of gangrene. He subsequently developed severe
CHF and was admitted to the nursing home as his wife could no longer take care of his
increasing physical needs at home.
The psychiatrist counseled the family and staff that Mr. M’s behaviors could be
managed in the long-term care facility if everyone collaborated in helping him. Family
and staff were informed that hospitalization carried its own risks of increased confusion,
delirium, functional decline, falls, and other iatrogenic problems.
The psychiatrist recommended starting sertraline 12.5 mg daily, increasing it every
seven days to a total of 50 mg daily. The psychiatrist also started Mr. M on clonazepam
0.25 mg in the morning and at bedtime but, due to daytime sedation, discontinued the
morning dose. The family was encouraged and successful in finding a World War II
veteran to visit Mr. M several times a week. Only comedy and game shows were kept on
the TV in his room, as well as in other places by astute staff. The psychiatrist also
recommended to the family and staff to avoid discussion of the terrorist attacks with
Mr. M and to have him avoid watching the news on TV. A list of topics for conversation
that did not involve war, politics, religion, or terrorist attacks was devised in consultation
with the staff and family.
After two weeks, Mr. M was less agitated but continued to have nightmares, aggres-
sion, and nighttime agitation. The psychiatrist discussed the use of prazosin for
158
“Bombs Are Falling! Run, Run!” 159
nightmares but, because of the significant risk of orthostatic hypotension, it was not
started. The psychiatrist increased the dosage of clonazepam to 0.5 mg at bedtime, but
Mr. M had two falls (no injuries) and the dose was decreased to 0.25 mg. The psychiatrist
added mirtazapine 7.5 mg at bedtime and Mr. M started sleeping better over the next two
weeks. Over the next three months, Mr. M gradually became significantly less anxious,
started sleeping better regularly, and his verbal and physical aggression resolved.
Teaching Points
Post-traumatic stress disorder is a mental health condition that is seen in patients who
have experienced or observed a traumatic event. Traumatic events are usually those
events that threaten death, serious injury, or sexual assault. Common traumatic experi-
ences that are known to sometimes lead to PTSD include war-related events, serious
accidents, natural disasters, physical or emotional abuse, serious illness, or the loss of
loved ones. Symptoms may include intrusive memories of the traumatic event, flash-
backs, nightmares, anxiety, and avoidance of reminders of the traumatic event. These
patients are also at increased risk of psychiatric comorbidities including depression and
anxiety. Pharmacologic treatments are often very helpful in lessening the symptoms of
PTSD. Nonpharmacologic interventions and support from family and caregivers are also
key to optimizing the quality of life in patients with PTSD.
Large epidemiologic studies have found that the lifetime prevalence of PTSD as
defined by DSM–5–TR criteria is 6%. Rates are generally higher in women (8%) than
in men (4%) [1], and veterans have a much higher prevalence. Lifetime prevalence was
3%, 10%, 21%, and 29% for those veterans that served in World War II/Korean War, the
Vietnam War, Persian Gulf War, and Operations Enduring Freedom and Iraqi Freedom,
respectively [2]. The declining prevalence with age may reflect that the symptoms of
PTSD often improve with time. Improved coping strategies with age may help older
adults manage the consequences of experiencing traumatic events more effectively.
The pathophysiology of PTSD is complex and exact mechanisms are still not fully
understood but are known to involve neurotransmitter imbalances, hyperactivity of the
amygdala, hippocampal changes, prefrontal cortex dysfunction, HPA axis dysregulation,
and neuroinflammation. See Box 32.1 for a more detailed description of these factors [3].
The development and maintenance of PTSD symptoms after a traumatic event are
influenced by key psychosocial factors. Understanding these factors can help to better
understand those with PTSD and design effective psychosocial interventions to help
them. Box 32.2 lists some psychosocial factors that are important in the development and
maintenance of PTSD [4, 5].
The diagnosis of PTSD is usually through a comprehensive evaluation by a mental
health professional, usually a psychiatrist, psychologist, or licensed therapist. The process
starts with a detailed assessment of the patient’s symptoms, medical history, and any
potential traumatic events that the patient has experienced. The DSM–5–TR criteria for
PTSD are listed in Box 32.3 [6].
There are some issues particular to the diagnosis of PTSD in older adults [7]. Older
adults may be less likely to discuss traumatic experiences due to the perceived stigma of
mental health disorders. Cognitive impairment may render older adults less able to
describe their symptoms or traumatic events. Older adults are more likely to have
medical or neurologic comorbidities with symptoms that can overlap with PTSD
160 Anxiety and Related Disorders
Box 32.2 Psychosocial factors that are important in the development and
maintenance of PTSD
1. Severity, nature, and duration of the traumatic event. Events involving physical harm,
sexual violence, or life-threatening situations are more likely to lead to PTSD.
Prolonged traumas are also more likely to lead to PTSD.
2. Perceived level of threat. A higher level of perceived threat can increase the likelihood
of developing and the severity of PTSD symptoms.
3. Social support. Those with adequate or good social support are less likely to develop
PTSD after a traumatic experience.
4. Coping mechanisms. Those individuals with good coping mechanisms, such as the
likelihood of seeking professional help, talking about experiences, and engaging in
relaxing behaviors, have improved outcomes. Avoiding emotions and relying on
maladaptive coping mechanisms such as substance abuse negatively affect outcomes.
5. Pre-existing mental health conditions. Those with pre-existing depression or anxiety
disorders are likely more vulnerable to developing PTSD after a traumatic event.
6. Personality traits. Certain personality traits, such as neuroticism or a history of
emotional instability, may increase the likelihood of developing and the severity of
PTSD symptoms.
7. Childhood factors. Those who have experienced childhood abuse or neglect may have
more trouble dealing with trauma later in life.
8. Post-trauma stress. Continued stress after a traumatic event can increase the likelihood
of severe or prolonged PTSD symptoms.
9. Avoidance behaviors. Avoidance of thoughts or discussions involving the traumatic
event can prevent emotional processing and hinder recovery.
symptoms. They may also have experienced multiple traumatic events during their
lifetime which can make identifying a specific event and how that event contributes to
the development of PTSD difficult. Grief and loss are common in later life and the
symptoms of complex grief can make it challenging to distinguish from PTSD symp-
toms. Diminished social support in older adults can impact their ability to seek help and
access mental health services.
“Bombs Are Falling! Run, Run!” 161
2. Presence of one (or more) of the following intrusion symptoms associated with the
traumatic event(s), beginning after the traumatic event(s) occurred:
a. Recurrent, involuntary, and intrusive distressing memories of the traumatic
event(s).
Note: In children older than six years, repetitive play may occur in which themes or
aspects of the traumatic event(s) are expressed.
b. Recurrent distressing dreams in which the content and/or effect of the dream are
related to the traumatic event(s).
Note: In children, there may be frightening dreams without recognizable content.
c. Dissociative reactions (e.g. flashbacks) in which the individual feels or acts as if the
traumatic event(s) were recurring. (Such reactions may occur on a continuum, with
the most extreme expression being a complete loss of awareness of present
surroundings.)
Note: In children, trauma-specific reenactment may occur in play.
d. Intense or prolonged psychological distress at exposure to internal or external cues
that symbolize or resemble an aspect of the traumatic event(s).
e. Marked physiological reactions to internal or external cues that symbolize or
resemble an aspect of the traumatic event(s).
3. Persistent avoidance of stimuli associated with the traumatic event(s), beginning after
the traumatic event(s) occurred, as evidenced by one or both of the following:
a. Avoidance of or efforts to avoid distressing memories, thoughts, or feelings about
or closely associated with the traumatic event(s).
b. Avoidance of or efforts to avoid external reminders (people, places, conversations,
activities, objects, situations) that arouse distressing memories, thoughts, or
feelings about or closely associated with the traumatic event(s).
4. Negative alterations in cognitions and mood associated with the traumatic event(s),
beginning or worsening after the traumatic event(s) occurred, as evidenced by two (or
more) of the following:
a. Inability to remember an important aspect of the traumatic event(s) (typically due to
dissociative amnesia and not to other factors such as head injury, alcohol, or drugs).
162 Anxiety and Related Disorders
5. Marked alterations in arousal and reactivity associated with the traumatic event(s),
beginning or worsening after the traumatic event(s) occurred, as evidenced by two (or
more) of the following:
a. Irritable behavior and angry outbursts (with little or no provocation) typically
expressed as verbal or physical aggression toward people or objects.
b. Reckless or self-destructive behavior.
c. Hypervigilance.
d. Exaggerated startle response.
e. Problems with concentration.
f. Sleep disturbance (e.g. difficulty falling or staying asleep or restless sleep).
Specify whether:
With dissociative symptoms: The individual’s symptoms meet the criteria for PTSD, and
in addition, in response to the stressor, the individual experiences persistent or recurrent
symptoms of either of the following:
1. Depersonalization: Persistent or recurrent experiences of feeling detached from,
and as if one were an outside observer of, one’s mental processes or body (e.g.
feeling as though one were in a dream; feeling a sense of unreality of self or body
or of time moving slowly).
2. Derealization: Persistent or recurrent experiences of unreality of surroundings
(e.g. the world around the individual is experienced as unreal, dreamlike, distant, or
distorted).
Note: To use this subtype, the dissociative symptoms must not be attributable to the
physiological effects of a substance (e.g. blackouts, behavior during alcohol intoxication)
or another medical condition (e.g. complex partial seizures).
Specify if:
With delayed expression: If the full diagnostic criteria are not met by at least six
months after the event (although the onset and expression of some symptoms may
be immediate).
“Bombs Are Falling! Run, Run!” 163
Take-Home Points
The pharmacologic treatment of PTSD attempts to alleviate the symptoms associated with the
condition including anxiety, depression, and sleep disturbances. SSRIs are first-line medications for
PTSD. SNRIs such as venlafaxine are also effective, especially in instances where there has been a
suboptimal response to SSRIs.
Follow-up for those with PTSD should involve regular visits with a provider to assess response to
treatment. Rating scales such as the PTSD Checklist 5 can be quite helpful in objectively assessing
the severity and nature of PTSD symptoms over time.
There are quite a few options for nonpharmacologic therapy in older adults. Outcomes are best in
those who participate in both pharmacologic and nonpharmacologic treatments. Some of the best
outcomes are seen with CBT combined with pharmacotherapy.
The prognosis of PTSD varies widely among individuals. Some patients may experience significant
improvement or even full remission of symptoms over time.
Ms. A was a 66-year-old female who was brought to her physician in the geriatric
psychiatry clinic by her caseworker to initiate care and for evaluation of “episodes”.
Ms. A was living on her own in a federally subsidized low-income senior apartment
building. Her caseworker provided support to her and others in her building. She had
Medicaid-funded chore worker support in the home for two hours per day, five days a
week. Ms. A had not driven in some years.
Ms. A was not very forthcoming with any psychiatric history. She said she was doing
“fine” and just wanted to be left alone. Her caseworker informed the psychiatrist that
Ms. A had not been doing very well in recent months and there was talk of her moving
into a long-term care facility due to concerns about appropriateness of her being by
herself. Ms. A had three emergency room visits in the previous four months, each after
falls and altered mental status/dehydration. A typical scenario would be the chore worker
finding Ms. A on the floor in her apartment, groggy and confused. Emergency medical
services would be called and the patient taken to the emergency room to be treated as an
inpatient for a day or two for dehydration or UTI.
Ms. A did not have much to add to her caseworker’s concerns other than to say that
she dreaded moving out of her apartment and did not see any reason to go to a nursing
home. When asked about her mood she stated that it was generally good but did admit to
some anxiety at times. Her PHQ-9 score was nine, indicating mild depression symptoms,
and her GAD–7 score was ten, indicating fairly mild anxiety. Her SLUMS exam score
was 24/30, indicating mild cognitive impairment. When Ms. A was asked what the
psychiatrist could do for her that might help her Ms. A replied, “Tell her [caseworker]
I can stay in my apartment.”
Ms. A’s only known psychiatric diagnosis was mild anxiety and depression for which
she took sertraline 100 mg as prescribed by her PCP. Her medical comorbidities included
hypothyroidism and hypertension. She did smoke cigarettes and had a history of drug
use. “I used to do a bit of everything,” she said and admitted to a history of abusing
alcohol, prescription pills, and methamphetamine mostly.
Her only living relative was a daughter who was serving time in prison for drug-
related charges and she had never been married. She lived on disability from the age of
46 and stated her disability was related to “a bad neck that really doesn’t bother me
anymore”. She had not graduated from high school. Before her approval for disability,
she had worked for some years in jobs such as patient care technician, waitress, and
convenience store attendant.
The psychiatrist probed Ms. A about her tendency to be hospitalized after being
found on the floor of her apartment, as well as being found on the floor in the stairwell of
167
168 Substance Abuse Disorders
her apartment building. The clinician told Ms. A that his goal was to help her stay as
independent as possible and try to assess the appropriateness of her staying in an
independent apartment.
When asked about her social life, Ms. A said she had few friends and preferred to
keep to herself. She had liked one or two of her chore workers in the past and considered
them to be the closest thing to friends that she had in the last several years. When asked
about hobbies she admitted to enjoying television and occasionally hanging out with a
new friend she had met in the neighborhood. As the interview progressed
Ms. A admitted she was smoking crack cocaine in the parking lot of her apartment.
Her caseworker appeared shocked and said, “You told me you didn’t do that anymore!”
to which Ms. A replied, “I don’t . . . only when I can afford it.”
As the history fleshed out it seemed that Ms. A was smoking crack cocaine in the
parking lot with a local dealer who would then help her back into the building. This was
happening once or twice per month as Ms. A informed the clinician that it cost her thirty
dollars and she was only able to afford that kind of expense once or twice a month at
most. If it were not for the cost, Ms. A stated she would use crack every day as it gave her
a rush of euphoria and “makes me forget about things”.
The psychiatrist and Ms. A’s caseworker emphasized to Ms. A that her crack cocaine
use was likely the cause of her “episodes” that required hospitalization and that her
continued use would likely jeopardize her ability to stay in the senior living apartment.
Ms. A refused to admit the crack use was a problem but did agree that she would try not
to use it so that she could stay in her apartment and avoid having to live in a
nursing home.
On her follow-up visit two months later, Ms. A was relatively unchanged. She had
another hospital admission where she presented with dehydration, at which time the
emergency room confirmed her urine was positive for cocaine. The caseworker worked
to find Ms. A a bed in a skilled nursing facility and, at a subsequent three-month follow-
up visit, Ms. A was brought to see her psychiatrist from such a facility where she had
been admitted for long-term care. She was doing well medically and had not had any
hospital visits. She was quite bitter about being in the long-term care facility but
admitted that some of the people were nice. She missed the freedom of being in
her apartment.
Teaching Points
As with younger adults, substance abuse among older adults is an increasing burden on
the healthcare system. Despite studies showing the increasing prevalence of substance
use disorders (SUD) in older adults, they remain underestimated, underdiagnosed, and
undertreated in this population. Identification of substance use problems is thought to be
more difficult as secondary signs of drug use such as job loss, relationship problems, or
legal issues may be lacking. Physical and cognitive declines thought secondary to drug
use can be easily misattributed to age-related cognitive or medical conditions. Primary
care providers and specialists alike may be less likely to probe into the possibilities of
drug use due to a lack of time or a misperception that illicit substance use is more of an
issue for younger patients [1]. Other misconceptions now questioned are that most illicit
drug use initiation occurs before the age of 30 and rarely occurs after. This may lead to
physicians screening less for these conditions in older adults.
“I Used to Do a Bit of Everything” 169
contribute to increased substance use. Interventions for opioid use disorders in older
adults can be similar to treatment in younger adults and can include medication-assisted
treatment (buprenorphine), psychotherapy, support groups, alternative pain manage-
ment plans, and increased social support to help with loneliness and social isolation.
The use of methamphetamines has increased alongside that of the use of other illicit
substances in recent years. Some parts of the country have experienced epidemic levels of
methamphetamine abuse, with contributing factors including wide availability due to
low cost and ease of manufacture. Methamphetamines are often used along with other
illicit substances to enhance the euphoric effects or mitigate the side effects of other
substances. They are highly addictive and medication treatment options are limited.
Rates of use are particularly high in lower socioeconomic groups who may have more
limited access to medical treatment. Besides addiction, chronic methamphetamine use
can lead to multiple health consequences including cognitive impairment, increased risk
of mood disorders, dental decay, malnutrition, and social and legal problems. The
epidemic of methamphetamine use in some areas of the country has not spared older
adults and a high index of suspicion should be maintained when evaluating older adults
with neuropsychiatric symptoms [9].
Take-Home Points
The potential of SUDs in older adults is often overlooked in a general health assessment. Substance
use disorders have a high comorbidity with other psychiatric disorders.
Cannabis use is increasing in all age groups, including older adults. Be aware that older adults may
be using cannabis to self-medicate psychiatric conditions such as anxiety and depression or to treat
chronic pain despite limited evidence for long-term improvement.
Psychological changes in older adults make them more susceptible to the negative effects of
alcohol use. With the proper support and resources, older adults with alcohol use disorder can live a
healthier, happier life free from alcohol.
Older adults may be at risk of opiate use disorder due to chronic pain issues, multiple medical
comorbidities, and psychiatric comorbidities. Treatment options for opioid use disorder, such as
medications, outpatient treatment programs, and psychosocial supports are often as effective in
older adults as in younger patients.
Ms. A was a 66-year-old female who was brought to her physician in the geriatric
psychiatry clinic by her caseworker to initiate care and for evaluation of “episodes”.
Ms. A was living on her own in a federally subsidized low-income senior apartment
building. Her caseworker provided support to her and others in her building. She had
Medicaid-funded chore worker support in the home for two hours per day, five days a
week. Ms. A had not driven in some years.
Ms. A was not very forthcoming with any psychiatric history. She said she was doing
“fine” and just wanted to be left alone. Her caseworker informed the psychiatrist that
Ms. A had not been doing very well in recent months and there was talk of her moving
into a long-term care facility due to concerns about appropriateness of her being by
herself. Ms. A had three emergency room visits in the previous four months, each after
falls and altered mental status/dehydration. A typical scenario would be the chore worker
finding Ms. A on the floor in her apartment, groggy and confused. Emergency medical
services would be called and the patient taken to the emergency room to be treated as an
inpatient for a day or two for dehydration or UTI.
Ms. A did not have much to add to her caseworker’s concerns other than to say that
she dreaded moving out of her apartment and did not see any reason to go to a nursing
home. When asked about her mood she stated that it was generally good but did admit to
some anxiety at times. Her PHQ-9 score was nine, indicating mild depression symptoms,
and her GAD–7 score was ten, indicating fairly mild anxiety. Her SLUMS exam score
was 24/30, indicating mild cognitive impairment. When Ms. A was asked what the
psychiatrist could do for her that might help her Ms. A replied, “Tell her [caseworker]
I can stay in my apartment.”
Ms. A’s only known psychiatric diagnosis was mild anxiety and depression for which
she took sertraline 100 mg as prescribed by her PCP. Her medical comorbidities included
hypothyroidism and hypertension. She did smoke cigarettes and had a history of drug
use. “I used to do a bit of everything,” she said and admitted to a history of abusing
alcohol, prescription pills, and methamphetamine mostly.
Her only living relative was a daughter who was serving time in prison for drug-
related charges and she had never been married. She lived on disability from the age of
46 and stated her disability was related to “a bad neck that really doesn’t bother me
anymore”. She had not graduated from high school. Before her approval for disability,
she had worked for some years in jobs such as patient care technician, waitress, and
convenience store attendant.
The psychiatrist probed Ms. A about her tendency to be hospitalized after being
found on the floor of her apartment, as well as being found on the floor in the stairwell of
167
168 Substance Abuse Disorders
her apartment building. The clinician told Ms. A that his goal was to help her stay as
independent as possible and try to assess the appropriateness of her staying in an
independent apartment.
When asked about her social life, Ms. A said she had few friends and preferred to
keep to herself. She had liked one or two of her chore workers in the past and considered
them to be the closest thing to friends that she had in the last several years. When asked
about hobbies she admitted to enjoying television and occasionally hanging out with a
new friend she had met in the neighborhood. As the interview progressed
Ms. A admitted she was smoking crack cocaine in the parking lot of her apartment.
Her caseworker appeared shocked and said, “You told me you didn’t do that anymore!”
to which Ms. A replied, “I don’t . . . only when I can afford it.”
As the history fleshed out it seemed that Ms. A was smoking crack cocaine in the
parking lot with a local dealer who would then help her back into the building. This was
happening once or twice per month as Ms. A informed the clinician that it cost her thirty
dollars and she was only able to afford that kind of expense once or twice a month at
most. If it were not for the cost, Ms. A stated she would use crack every day as it gave her
a rush of euphoria and “makes me forget about things”.
The psychiatrist and Ms. A’s caseworker emphasized to Ms. A that her crack cocaine
use was likely the cause of her “episodes” that required hospitalization and that her
continued use would likely jeopardize her ability to stay in the senior living apartment.
Ms. A refused to admit the crack use was a problem but did agree that she would try not
to use it so that she could stay in her apartment and avoid having to live in a
nursing home.
On her follow-up visit two months later, Ms. A was relatively unchanged. She had
another hospital admission where she presented with dehydration, at which time the
emergency room confirmed her urine was positive for cocaine. The caseworker worked
to find Ms. A a bed in a skilled nursing facility and, at a subsequent three-month follow-
up visit, Ms. A was brought to see her psychiatrist from such a facility where she had
been admitted for long-term care. She was doing well medically and had not had any
hospital visits. She was quite bitter about being in the long-term care facility but
admitted that some of the people were nice. She missed the freedom of being in
her apartment.
Teaching Points
As with younger adults, substance abuse among older adults is an increasing burden on
the healthcare system. Despite studies showing the increasing prevalence of substance
use disorders (SUD) in older adults, they remain underestimated, underdiagnosed, and
undertreated in this population. Identification of substance use problems is thought to be
more difficult as secondary signs of drug use such as job loss, relationship problems, or
legal issues may be lacking. Physical and cognitive declines thought secondary to drug
use can be easily misattributed to age-related cognitive or medical conditions. Primary
care providers and specialists alike may be less likely to probe into the possibilities of
drug use due to a lack of time or a misperception that illicit substance use is more of an
issue for younger patients [1]. Other misconceptions now questioned are that most illicit
drug use initiation occurs before the age of 30 and rarely occurs after. This may lead to
physicians screening less for these conditions in older adults.
“I Used to Do a Bit of Everything” 169
contribute to increased substance use. Interventions for opioid use disorders in older
adults can be similar to treatment in younger adults and can include medication-assisted
treatment (buprenorphine), psychotherapy, support groups, alternative pain manage-
ment plans, and increased social support to help with loneliness and social isolation.
The use of methamphetamines has increased alongside that of the use of other illicit
substances in recent years. Some parts of the country have experienced epidemic levels of
methamphetamine abuse, with contributing factors including wide availability due to
low cost and ease of manufacture. Methamphetamines are often used along with other
illicit substances to enhance the euphoric effects or mitigate the side effects of other
substances. They are highly addictive and medication treatment options are limited.
Rates of use are particularly high in lower socioeconomic groups who may have more
limited access to medical treatment. Besides addiction, chronic methamphetamine use
can lead to multiple health consequences including cognitive impairment, increased risk
of mood disorders, dental decay, malnutrition, and social and legal problems. The
epidemic of methamphetamine use in some areas of the country has not spared older
adults and a high index of suspicion should be maintained when evaluating older adults
with neuropsychiatric symptoms [9].
Take-Home Points
The potential of SUDs in older adults is often overlooked in a general health assessment. Substance
use disorders have a high comorbidity with other psychiatric disorders.
Cannabis use is increasing in all age groups, including older adults. Be aware that older adults may
be using cannabis to self-medicate psychiatric conditions such as anxiety and depression or to treat
chronic pain despite limited evidence for long-term improvement.
Psychological changes in older adults make them more susceptible to the negative effects of
alcohol use. With the proper support and resources, older adults with alcohol use disorder can live a
healthier, happier life free from alcohol.
Older adults may be at risk of opiate use disorder due to chronic pain issues, multiple medical
comorbidities, and psychiatric comorbidities. Treatment options for opioid use disorder, such as
medications, outpatient treatment programs, and psychosocial supports are often as effective in
older adults as in younger patients.
Ms. G was an 82-year-old female. She had recently moved into long-term care after a stay
in rehab following a fall at home and a left hip fracture. She was now confined to a
wheelchair and needed assistance with many ADLs such as showering, dressing, toileting,
and incontinence care. She was cognitively intact and could direct her care but had a
difficult time adjusting to her new home in long-term care. In the short time she had
been a resident at the facility she had shown a tendency toward not getting along with
others, including care staff. Some staff had gone as far as to refuse to work an assigned
shift if they knew it would involve caring for Ms. G. She was described as demanding,
hypercritical, demeaning to others, and snobbish. Staff asked her provider to evaluate her
for a potential untreated psychiatric condition that could be making it difficult for
Ms. G to fit in at the facility.
On the initial visit with Ms. G, she was found in her room dressed in a fluffy
bathrobe, a large string of pearls, large earrings, and rings on every finger of a costume
jewelry style. The walls of her room were covered with photos of herself, and several of
were of the “glamour shot” style that was popular in the mid-1990s. Ms. G’s greeting was
warm and almost before introductions were finished, she delved into a 30-minute
monologue describing the accomplishments of her life and her associations with many
important people in the community. Attempts to interrupt Ms. G by the provider were
greeted with acidic irritation. The provider listened quietly for some time, largely out of
fascination for the outlandishness of the narrative that unfolded. Ms. G described a life
that was full of meaningful and close relationships with many successful people in all
areas including politics, the arts, and business. She described her talents in business, the
arts, and later-life philanthropic ventures in great detail.
When asked about her family, Ms. G admitted to having three children who she
described as “wildly successful”. She had married three times and divorced three times.
When asked if the provider could speak with one of her children, Ms. G stated that they
were all “terribly busy” and would likely not have time to speak with a doctor. When
asked about the facility and her adjustment to it she had little good to say. She described
the perceived incompetencies of the staff, the inedible nature of the food, and the drab
decor. She mentioned she was at the facility only long enough for her to find accommo-
dations she was more accustomed to. When asked about activities she enjoyed she
mentioned that she found none of the activities enjoyable as none of the other residents
were “on my level”.
Ms. G was on no psychiatric medications. She stated she had no history of mental
health issues and had never been hospitalized for mental health reasons. She had no
history of drug or alcohol abuse but admitted that she needed a good martini every
173
174 Personality Disorders
evening to “stay sane” and was furious that there was not a bartender in the facility who
could accommodate her.
Ms. G’s SLUMS score was 26. She showed no outward signs of clear cognitive
impairment although she was at times hard to keep on topic.
The clinician called Ms. G’s youngest daughter who was listed as her power of
attorney. The daughter was quite willing to discuss her mother’s psychiatric history
and stated that Ms. G had been a “difficult woman” her entire life. Her life was dotted
with troubled relationships; she had been married three times and each divorce was non-
amicable. She tended to alienate any new friends she would make after only a short time.
During recent years she began needing more and more help at home and her daughter
had personally arranged for a total of eight different caregivers in a year who each quit –
usually after a matter of days or weeks due to her mother’s demanding nature and queen-
like attitude. Ms. G’s daughter encouraged the provider to believe less than half of what
Ms. G said as she greatly exaggerated her achievements and the perceived incompetencies
of others. Ms. G was moved into the nursing home as her family could find no others to
care for her at home.
After a long conversation with the daughter and conversations with care staff at the
facility the long-term care provider felt that Ms. G met no criteria for any clinical mood
or anxiety disorders. She did not appear to have more than mild cognitive dysfunction
and had no signs of any psychosis. The pattern of current and past relationship
dysfunction suggested a diagnosis of narcissistic personality disorder. Staff were
informed of the provider’s opinion on the patient’s mental health diagnosis and gave
them strategies to better deal with the challenging behaviors of Ms. G. The provider also
recommended regular psychotherapy to focus on potential ways that she could improve
her interactions with others and make for a more pleasurable living situation for her and
a better work environment for those who helped care for her.
Teaching Points
Personality traits are patterns of thoughts, feelings, and behaviors that remain relatively
stable over a person’s life and strongly influence how they interact with and perceive the
world. These traits can be adaptive and lead to improved psychosocial function or be
maladaptive and lead to impairment in psychosocial function. When personality traits
come together in particular maladaptive patterns and lead to dysfunction and significant
impairment in personal, family, social, educational, occupational, or other important
areas of functioning they can be classified as a personality disorder. Personality disorders
are relatively common. Estimates are that about 12% of the general population, 25% of
primary care patients, and at least 50% of psychiatric outpatients meet the criteria for a
personality disorder [1].
Those with personality disorders usually show signs of personality traits and behav-
ioral patterns in adolescence that can later solidify into a recognized personality disorder
in late adolescence or early adulthood.
The DSM–5 lists ten distinct personality disorders, which can be classified into three
subgroups. Cluster A personality disorders are characterized by traits and behaviors that
are often seen as odd or eccentric and these patients may have difficulty with social
relationships due to their unusual behaviors and thought patterns. Cluster A personality
disorders include paranoid personality disorder, schizoid personality disorder, and
“No One Here Is on My Level” 175
Take-Home Points
Personality traits are patterns of thoughts, feelings, and behaviors that remain relatively stable over
a person’s life and strongly influence how people interact with and perceive the world.
Personality disorders are personality traits that cluster in recognizable patterns and produce
dysfunction in multiple areas of life.
Personality disorders do not respond well to pharmacologic interventions but are often accompan-
ied by other psychiatric comorbidities that can be treated medically.
Recognizing personality disorders can help a provider avoid misdiagnosing psychiatric conditions,
anticipate future care challenges, and better counsel family and caregivers.
“No One Here Is on My Level” 177
Ms. G was an 82-year-old female. She had recently moved into long-term care after a stay
in rehab following a fall at home and a left hip fracture. She was now confined to a
wheelchair and needed assistance with many ADLs such as showering, dressing, toileting,
and incontinence care. She was cognitively intact and could direct her care but had a
difficult time adjusting to her new home in long-term care. In the short time she had
been a resident at the facility she had shown a tendency toward not getting along with
others, including care staff. Some staff had gone as far as to refuse to work an assigned
shift if they knew it would involve caring for Ms. G. She was described as demanding,
hypercritical, demeaning to others, and snobbish. Staff asked her provider to evaluate her
for a potential untreated psychiatric condition that could be making it difficult for
Ms. G to fit in at the facility.
On the initial visit with Ms. G, she was found in her room dressed in a fluffy
bathrobe, a large string of pearls, large earrings, and rings on every finger of a costume
jewelry style. The walls of her room were covered with photos of herself, and several of
were of the “glamour shot” style that was popular in the mid-1990s. Ms. G’s greeting was
warm and almost before introductions were finished, she delved into a 30-minute
monologue describing the accomplishments of her life and her associations with many
important people in the community. Attempts to interrupt Ms. G by the provider were
greeted with acidic irritation. The provider listened quietly for some time, largely out of
fascination for the outlandishness of the narrative that unfolded. Ms. G described a life
that was full of meaningful and close relationships with many successful people in all
areas including politics, the arts, and business. She described her talents in business, the
arts, and later-life philanthropic ventures in great detail.
When asked about her family, Ms. G admitted to having three children who she
described as “wildly successful”. She had married three times and divorced three times.
When asked if the provider could speak with one of her children, Ms. G stated that they
were all “terribly busy” and would likely not have time to speak with a doctor. When
asked about the facility and her adjustment to it she had little good to say. She described
the perceived incompetencies of the staff, the inedible nature of the food, and the drab
decor. She mentioned she was at the facility only long enough for her to find accommo-
dations she was more accustomed to. When asked about activities she enjoyed she
mentioned that she found none of the activities enjoyable as none of the other residents
were “on my level”.
Ms. G was on no psychiatric medications. She stated she had no history of mental
health issues and had never been hospitalized for mental health reasons. She had no
history of drug or alcohol abuse but admitted that she needed a good martini every
173
174 Personality Disorders
evening to “stay sane” and was furious that there was not a bartender in the facility who
could accommodate her.
Ms. G’s SLUMS score was 26. She showed no outward signs of clear cognitive
impairment although she was at times hard to keep on topic.
The clinician called Ms. G’s youngest daughter who was listed as her power of
attorney. The daughter was quite willing to discuss her mother’s psychiatric history
and stated that Ms. G had been a “difficult woman” her entire life. Her life was dotted
with troubled relationships; she had been married three times and each divorce was non-
amicable. She tended to alienate any new friends she would make after only a short time.
During recent years she began needing more and more help at home and her daughter
had personally arranged for a total of eight different caregivers in a year who each quit –
usually after a matter of days or weeks due to her mother’s demanding nature and queen-
like attitude. Ms. G’s daughter encouraged the provider to believe less than half of what
Ms. G said as she greatly exaggerated her achievements and the perceived incompetencies
of others. Ms. G was moved into the nursing home as her family could find no others to
care for her at home.
After a long conversation with the daughter and conversations with care staff at the
facility the long-term care provider felt that Ms. G met no criteria for any clinical mood
or anxiety disorders. She did not appear to have more than mild cognitive dysfunction
and had no signs of any psychosis. The pattern of current and past relationship
dysfunction suggested a diagnosis of narcissistic personality disorder. Staff were
informed of the provider’s opinion on the patient’s mental health diagnosis and gave
them strategies to better deal with the challenging behaviors of Ms. G. The provider also
recommended regular psychotherapy to focus on potential ways that she could improve
her interactions with others and make for a more pleasurable living situation for her and
a better work environment for those who helped care for her.
Teaching Points
Personality traits are patterns of thoughts, feelings, and behaviors that remain relatively
stable over a person’s life and strongly influence how they interact with and perceive the
world. These traits can be adaptive and lead to improved psychosocial function or be
maladaptive and lead to impairment in psychosocial function. When personality traits
come together in particular maladaptive patterns and lead to dysfunction and significant
impairment in personal, family, social, educational, occupational, or other important
areas of functioning they can be classified as a personality disorder. Personality disorders
are relatively common. Estimates are that about 12% of the general population, 25% of
primary care patients, and at least 50% of psychiatric outpatients meet the criteria for a
personality disorder [1].
Those with personality disorders usually show signs of personality traits and behav-
ioral patterns in adolescence that can later solidify into a recognized personality disorder
in late adolescence or early adulthood.
The DSM–5 lists ten distinct personality disorders, which can be classified into three
subgroups. Cluster A personality disorders are characterized by traits and behaviors that
are often seen as odd or eccentric and these patients may have difficulty with social
relationships due to their unusual behaviors and thought patterns. Cluster A personality
disorders include paranoid personality disorder, schizoid personality disorder, and
“No One Here Is on My Level” 175
Take-Home Points
Personality traits are patterns of thoughts, feelings, and behaviors that remain relatively stable over
a person’s life and strongly influence how people interact with and perceive the world.
Personality disorders are personality traits that cluster in recognizable patterns and produce
dysfunction in multiple areas of life.
Personality disorders do not respond well to pharmacologic interventions but are often accompan-
ied by other psychiatric comorbidities that can be treated medically.
Recognizing personality disorders can help a provider avoid misdiagnosing psychiatric conditions,
anticipate future care challenges, and better counsel family and caregivers.
“No One Here Is on My Level” 177
35 Miserable Place”
Borderline Personality Disorder
Ms. L was a 64-year-old woman admitted to a nursing home for rehabilitation after a
prolonged hospital stay (three months) for nephrectomy and post-nephrectomy compli-
cations (infection, bleeding, and delirium with agitation). The long-term care provider
was asked to help manage the resident’s hostile and aggressive behavior toward staff.
The staff expressed anger and resentment at having to provide personal care for
Ms. L because of her persistent name-calling, yelling at the staff, and demeaning them
almost daily. Some staff members had started to call in sick if they found out that they
had to care for Ms. L, and some refused to care for her.
The provider found Ms. L to be an intelligent person who had relatively intact
cognitive function (SLUMS score 26) and was quick to take offense but also willing to
accept help from “competent” professionals. She repeatedly expressed, “I cannot wait to
get out of this miserable place.”
Ms. L was taking sertraline, 100 mg daily, for MDD. After interviewing Ms. L, the
provider met with the staff to explain that, in his opinion, Ms. L had been experiencing
moderate to severe depression (MDD) (PHQ–9 score was 20) with an underlying long-
standing personality disorder. Ms. L had confided in the provider that she had been
severely physically and emotionally abused as a child and had experienced three “hor-
rible” marriages. She had expressed to the provider that she “hated everyone,” liked
animals more than people, had lots of pets that she took care of, and helped the Humane
Society in caring for sick pets.
The provider provided emotional support to the staff, validated their feelings related
to dealing with Ms. L’s negative behaviors, discussed specific therapeutic ways the staff
could respond to these negative behaviors, and explained the treatment plan (increase in
antidepressants and individual psychotherapy). The provider (with Ms. L’s permission)
shared minor details about her abusive childhood and spousal relationships and the good
volunteer work she was doing with the Humane Society. This helped the staff feel
supported, improved their compassion for Ms. L, and enabled them to be more patient
with her so that treatment could be given time to help.
The long-term care provider increased the dosage of sertraline from 100 to 150 mg
and encouraged the staff to tell Ms. L that she was being disrespectful when she used foul
language or yelled at them. Staff was also encouraged to respond calmly (without
resentment) and appreciatively when Ms. L talked respectfully. The provider gently
helped Ms. L understand that her hostile behavior put her at a much higher risk of
prolonged stay in the nursing home because the staff would be less likely to meet her
needs diligently or promptly. Ms. L agreed to go to an outpatient clinic once a week for
counseling with a social worker. Over the next few weeks, the frequency and intensity of
178
“I Cannot Wait to Get out of This Miserable Place” 179
hostile comments by Ms. L decreased modestly, the staff was able to meet more of her
needs, and Ms. L made significant progress in physical therapy.
Teaching Points
Depressive disorders are common among residents who have personality disorders and
are often underrecognized and undertreated. A resident who has a personality disorder
also generates strong negative feelings in others that may result in staff being less likely to
meet the resident’s needs. Interventions should be designed with the intention of making
at least modest changes in behavior possible to achieve a desired result. Interventions
may include pharmacotherapy for comorbid mood or anxiety disorder, staff education,
or counseling for the patient. Behavior goals may include less name-calling by the
resident toward staff, resulting in an increased likelihood that staff will be able to meet
the resident’s needs.
Psychotherapy is useful for residents who have personality disorders and relatively
intact cognitive function. Optimization of treatment of comorbid mental disorder (MDD
in this situation) and specific guidance for staff of therapeutic ways to respond to
negative behaviors are also crucial for a successful outcome.
Borderline personality disorder (BPD) is a complex mental health condition. Those
with the condition have unstable moods, a history of difficulty functioning in relation-
ships, and a dysfunctional self-image. Patients with BPD have emotional dysregulation
that can lead to impulsive behaviors, self-harm, and fear of abandonment. They also
struggle with chronic feelings of emptiness and loneliness. Caring for those with BPD
can be particularly challenging. Knowledge of the condition by healthcare professionals
can provide insight into patients suffering from BPD and help providers have more
compassionate and therapeutic interactions with them.
The prevalence of BPD is estimated to be between 1 and 6% [1]. Those with
diagnosed BPD are more likely to be female (75%) than male (25%) [2]. The symptoms
are usually recognized first in late adolescence or early adulthood, but it is also possible
to be diagnosed in late adulthood. Those with BPD use a disproportionately high amount
of psychiatric services and are more likely to have a history of multiple psychiatric
hospital admissions [3]. Studies also show that those with BPD are more likely to have
comorbid mood disorders.
The causes of BPD are complex. As with many mental health conditions, there is an
interplay between genetics and environmental factors. Although no specific gene or
genes have been identified, it is known that BPD tends to run in families. Individuals
with BPD have been shown to have a higher incidence of structural and functional
abnormalities of regions of the brain involved in emotional regulation and impulse
control. Those who have had childhood trauma, abuse, or neglect are more likely to
develop BPD.
Those with BPD have significant challenges in life that can lead to poor psychosocial
outcomes. Interpersonal relationships can often be intense and dysfunctional and dem-
onstrate frequent conflicts. Those with BPD may have a hard time maintaining long-
term relationships. Emotional dysregulation can lead to rapid and intense mood swings,
which often makes functioning in work environments very difficult. Impulsivity can lead
to issues such as reckless spending, risky sexual behaviors, and substance abuse. Those
with BPD are less likely to achieve educational goals, more likely to have legal issues, and
180 Personality Disorders
have less access to consistent treatment. Those with BPD are also at much higher risk of
suicide than the general population [4].
The diagnosis of BPD is usually made by a psychiatrist, psychologist, or other mental
health professional. The DSM–5–R criteria for the diagnosis of BPD are listed in
Box 35.1 [2].
The treatment of BPD is largely through long-term psychological therapy and the
gold standard approach is dialectical behavior therapy (DBT). This therapy focuses on
optimizing emotional regulation, distress tolerance, interpersonal effectiveness, and
mindfulness skills. Other therapies that have been used with success are CBT, schema-
focused therapy, and supportive psychotherapy, among others. There is no specific
medication approved for the treatment of BPD but those who have it often have
comorbid anxiety or depression that require pharmacologic interventions. Research
has suggested that approximately half of people who receive psychotherapy do not
respond to the treatment independent of psychotherapy modality or length of treatment
[5]. Issues with access to appropriately trained therapists are also widespread.
There are currently no FDA-approved medications for the treatment of BPD. Off-
label use of medications is common. A list of medication classes sometimes used to treat
BPD-associated symptoms is shown in Box 35.2 [6].
Interactions between healthcare providers and those with BPD can be challenging
and patients in the long-term care setting are at risk of poor outcomes as they are often
perceived as unpleasant and demanding. Setting boundaries is important when dealing
with these patients. Regular empathetic validation of feelings by the provider can
strengthen the therapeutic relationship between those with BPD and their providers.
The prognosis for borderline personality is generally mixed. Thankfully, a majority of
patients with BPD have a decline in symptoms during adulthood. It is estimated that up
“I Cannot Wait to Get out of This Miserable Place” 181
to 85% of patients with BPD reach a diagnostic remission within ten years of diagnosis.
Some residual symptoms may persist, leading to lifelong challenges. These may include
fear of abandonment, impulsivity, intense anger issues, and an unstable self-image that
can lead to lifelong social and occupational challenges [7]. Those with persistent BPD do
have a higher risk of death from suicide and medical comorbidities. The clinical course is
often marked by multiple suicidal attempts and up to 10% of those with BPD will die by
suicide. Common practice is still to admit those patients who make suicidal threats
despite the lack of evidence that hospitalization reduces the risk of actual suicide. There
is also no evidence that pharmacologic interventions reduce the risk of suicide in BPD
patients although they are effective in reducing the symptoms of comorbid mood or
anxiety symptoms [8].
The nature of BPD is generally chronic and persistent dysfunction, but the level of
dysfunction may actually decline with age. It is thought that age and maturity may
mellow the symptoms of the condition over the course of a lifetime. Cumulative
treatment and support can have a slow, steady, and positive effect in helping those with
BPD to manage their symptoms more effectively. The stable environment that a sup-
portive long-term care setting can provide may also have a positive impact on the
outcomes of older adults with BPD.
Therapeutic relationships between providers and those with BPD can be challenging.
A better understanding of the etiologic factors, treatment interventions, and comorbid-
ities can foster more empathy and understanding of these patients and allow the provider
to guide the treatment team in the long-term care setting more effectively.
Take-Home Points
Borderline personality disorder is a complex mental health condition. Those with the condition
have unstable moods, a history of difficulty functioning in relationships, and a dysfunctional self-
image. Patients with BPD have emotional dysregulation that can lead to impulsive behaviors, self-
harm, and fear of abandonment.
The treatment of BPD is largely through long-term psychological therapy and the gold standard
approach is DBT. This therapy focuses on optimizing emotional regulation, distress tolerance,
interpersonal effectiveness, and mindfulness skills.
Those with BPD have significant challenges in life that can lead to poor psychosocial outcomes.
Interpersonal relationships can often be intense and dysfunctional and demonstrate frequent
182 Personality Disorders
conflicts. Emotional dysregulation can lead to rapid and intense mood swings. Impulsivity can lead
to issues such as reckless spending, risky sexual behaviors, and substance abuse.
The nature of BPD is generally chronic and persistent dysfunction, but the level of dysfunction may
decline with age. It is thought that age and maturity may mellow the symptoms of the condition
over the course of a lifetime. Cumulative treatment and support can have a slow, steady, and
positive effect in helping those with BPD to manage their symptoms more effectively.
Mrs. M was a 91-year-old female who was a resident of a skilled nursing facility in a rural
area served via virtual visits from her geriatric psychiatrist. She had a history of
Alzheimer’s disease with behavioral disturbances, anxiety, insomnia, peripheral vascular
disease, adrenal insufficiency, and osteoporosis. Her cognitive impairment was moderate
to severe and she needed assistance with most ADLs. Mrs. M. was described as very
sociable and had been nicknamed “The Hostess” by staff as she liked to attend to other
residents in the unit and make sure they were comfortable. Sometimes she was confused
and perturbed as she felt that she was at home and that the other residents were guests
who would not leave, and she expressed fears that she would not be able to cover the cost
of the food and entertainment.
She was also described by staff as “a very sexual lady”. Her sexual behaviors had
become increasingly problematic over the course of weeks to months. Staff noted that
Mrs. M had lost her modesty, as demonstrated by often leaving her room without any
underwear or pants on. She was also noted to often be masturbating in common areas.
When attempts were made to redirect her to a more appropriate and private place she
would become angry and also verbally and physically aggressive to staff or other
residents. She had been noted to be more frequently touching male staff and residents
and had also been found trying to crawl into bed with other male residents. Staff brought
up these behaviors with the patient’s family but they found it hard to accept that their
mother could be doing these things as she had always been very proper and modest as a
younger adult.
The patient’s mood was described as mostly pleasant and content much of the time,
but she had been having increasing episodes of acting sad, anxious, or restless. During
the virtual interview with the patient, she was calm, pleasant, and was appropriately and
modestly dressed. She was unable to give any history due to her confusion and was
oriented to person only.
Her medication list was relatively short and included a multivitamin daily, hydrocor-
tisone tab 10 mg daily, acetaminophen 650 mg twice daily, quetiapine 25 mg daily,
melatonin 20 mg qhs, and hydroxyzine 25 mg twice daily as needed for anxiety.
A medical workup including blood work and urine tests was unrevealing. Her
psychiatrist decided to discontinue hydroxyzine due to anticholinergic effects in older
adults, her nightly melatonin dose was decreased from 20 to 5 mg, and her quetiapine
dose was increased from 25 g daily to 25 mg twice daily in an attempt to help her anxiety
and restlessness in the setting of advanced cognitive impairment. She was also started on
duloxetine 30 mg daily as her psychiatrist felt that she was showing signs of depression
and anxiety with an increase in agitation and restlessness.
183
184 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
At a follow-up appointment four weeks later, nursing staff reported that she was
possibly engaging in less sexually inappropriate behaviors than a month prior but the
behaviors were still thought to be problematic. Her duloxetine was then increased to
60 mg daily and nursing staff reported that the patient’s behaviors were much improved
at the subsequent four-week follow-up visit. She was still engaging in occasional mastur-
bation in common areas but was easily redirected to her room. She was generally much
less irritable and anxious. It was decided to continue the low-dose quetiapine and the
60 mg daily dose of duloxetine.
Teaching Points
This is an interesting case on several fronts. It brings into focus issues involving sexuality
and sexual expression in older adults with cognitive impairment.
Disinhibited behavior is commonly demonstrated in seniors with MNCD. This can
be demonstrated by such things as pilfering, swearing, physical aggression, diminished
social graces, and unwanted sexual expression. Prevalence of sexually inappropriate
behavior (SIB) varies depending on the type of MNCD, but reports have shown rates
of 2–25% depending on the setting. Behavioral variant FTD patients likely have the
highest prevalence of SIB with estimates ranging from 8% to 18% [1]. Some examples of
SIB that can be demonstrated by those with MNCD are listed in Box 36.1.
It is a misconception that all those with MNCD lose the desire for sexual touch and
sexual expression as their disease progresses from the mild to more moderate or severe
stages of the disease. As with those younger patients unaffected by MNCD, the spectrum
of “normal” sexual behavior is wide and undoubtedly influenced by a patient’s under-
lying personality or past sexual experiences. Those with advanced MNCD may even
confuse those they encounter in day-to-day life with a spouse or significant other that has
passed away.
Sexual expression in those with MNCD therefore should not be universally discour-
aged, shamed, or medically treated as an aberrant behavior. Sexual expression such as
consensual physical touch, holding hands, and kissing can be essential to mental health
for those with MNCD who have the desire.
The workup of SIB in long-term care residents starts with a good history, often
obtained from caregivers. It is important to assess the context of SIB by answering
questions such as (1) what exactly is the behavior being demonstrated? (2) what is the
context of the behavior? (3) how often is the behavior occurring? (4) who is being
impacted by the behavior? (5) is the sexual behavior greatly out of proportion to other
disinhibited behaviors? (Figure 36.1).
Pharmacologic management strategies of SIB in those with MNCD are not predict-
able as there are no randomized controlled trials to establish the efficacy or safety of
various medication interventions. A good general approach to management involves first
addressing modifiable environmental factors that could be leading to SIB. Secondly,
medications known to possibly increase changes in SIB should be identified and removed
if possible (benzodiazepines, dopamine agonists). Finally, pharmacologic treatments
specific to the SIB can be trialed. Antidepressants such as SSRIs, trazodone, and
mirtazapine have been used with some success and are generally well tolerated.
Antiandrogens such as medroxyprogesterone, estrogen, and finasteride can be used in
men and have shown benefit. Anticonvulsants such as gabapentin and tegretol have been
used for SIB (Figure 36.2).
In the case presented here, the patient’s behavior was considered problematic as her
sexual behavior was in common areas among other staff and residents. Other residents
were uncomfortable. The dignity of the patient could not be ensured with sexual
behaviors occurring in public places.
Of interest to the psychiatrist was the fact that these behaviors seemed to escalate
in the setting of Mrs. M’s increasing outward signs of depression and anxiety.
Although studies specific to older adult patients are lacking, it is known that anxiety
and depression have been linked to hypersexual behavior. Also, anxiety and depres-
sion are the most common mental health diagnoses in those who could be considered
to have dysfunctional sexual behaviors. It was reasoned that the patient’s dysfunc-
tional sexual expression could be one of her manifestations of depression or anxiety.
This was the impetus to initiate more aggressive treatment with an SNRI/atypical
antipsychotic combination. Staff were encouraged to continue attempts to redirect
Mrs. M to private areas of the facility where she could express herself without
offending others and while maintaining her dignity. It was hoped that as the treatment
of her potential underlying mood disorder took effect, she would be less angry and
upset with attempts at redirection.
“Can I Have a Hug?” 187
Take-Home Points
Sexually inappropriate behavior can be a manifestation of disinhibited behavior seen in older adults
with MNCD.
The evaluation of SIB should include a detailed history of what the behavior is, who is involved,
what the context of the behavior is, who is being impacted, and if the behavior is out of proportion
with other disinhibited behaviors.
Sexual expression in older adults with MNCD is normal and should not be universally discouraged
when in the appropriate setting and when the rights of others are not violated.
Look for and potentially remove medications that could be contributing to SIB including dopamine
agonists, anticholinergic agents, and benzodiazepines.
Mrs. M was a 91-year-old female who was a resident of a skilled nursing facility in a rural
area served via virtual visits from her geriatric psychiatrist. She had a history of
Alzheimer’s disease with behavioral disturbances, anxiety, insomnia, peripheral vascular
disease, adrenal insufficiency, and osteoporosis. Her cognitive impairment was moderate
to severe and she needed assistance with most ADLs. Mrs. M. was described as very
sociable and had been nicknamed “The Hostess” by staff as she liked to attend to other
residents in the unit and make sure they were comfortable. Sometimes she was confused
and perturbed as she felt that she was at home and that the other residents were guests
who would not leave, and she expressed fears that she would not be able to cover the cost
of the food and entertainment.
She was also described by staff as “a very sexual lady”. Her sexual behaviors had
become increasingly problematic over the course of weeks to months. Staff noted that
Mrs. M had lost her modesty, as demonstrated by often leaving her room without any
underwear or pants on. She was also noted to often be masturbating in common areas.
When attempts were made to redirect her to a more appropriate and private place she
would become angry and also verbally and physically aggressive to staff or other
residents. She had been noted to be more frequently touching male staff and residents
and had also been found trying to crawl into bed with other male residents. Staff brought
up these behaviors with the patient’s family but they found it hard to accept that their
mother could be doing these things as she had always been very proper and modest as a
younger adult.
The patient’s mood was described as mostly pleasant and content much of the time,
but she had been having increasing episodes of acting sad, anxious, or restless. During
the virtual interview with the patient, she was calm, pleasant, and was appropriately and
modestly dressed. She was unable to give any history due to her confusion and was
oriented to person only.
Her medication list was relatively short and included a multivitamin daily, hydrocor-
tisone tab 10 mg daily, acetaminophen 650 mg twice daily, quetiapine 25 mg daily,
melatonin 20 mg qhs, and hydroxyzine 25 mg twice daily as needed for anxiety.
A medical workup including blood work and urine tests was unrevealing. Her
psychiatrist decided to discontinue hydroxyzine due to anticholinergic effects in older
adults, her nightly melatonin dose was decreased from 20 to 5 mg, and her quetiapine
dose was increased from 25 g daily to 25 mg twice daily in an attempt to help her anxiety
and restlessness in the setting of advanced cognitive impairment. She was also started on
duloxetine 30 mg daily as her psychiatrist felt that she was showing signs of depression
and anxiety with an increase in agitation and restlessness.
183
184 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
At a follow-up appointment four weeks later, nursing staff reported that she was
possibly engaging in less sexually inappropriate behaviors than a month prior but the
behaviors were still thought to be problematic. Her duloxetine was then increased to
60 mg daily and nursing staff reported that the patient’s behaviors were much improved
at the subsequent four-week follow-up visit. She was still engaging in occasional mastur-
bation in common areas but was easily redirected to her room. She was generally much
less irritable and anxious. It was decided to continue the low-dose quetiapine and the
60 mg daily dose of duloxetine.
Teaching Points
This is an interesting case on several fronts. It brings into focus issues involving sexuality
and sexual expression in older adults with cognitive impairment.
Disinhibited behavior is commonly demonstrated in seniors with MNCD. This can
be demonstrated by such things as pilfering, swearing, physical aggression, diminished
social graces, and unwanted sexual expression. Prevalence of sexually inappropriate
behavior (SIB) varies depending on the type of MNCD, but reports have shown rates
of 2–25% depending on the setting. Behavioral variant FTD patients likely have the
highest prevalence of SIB with estimates ranging from 8% to 18% [1]. Some examples of
SIB that can be demonstrated by those with MNCD are listed in Box 36.1.
It is a misconception that all those with MNCD lose the desire for sexual touch and
sexual expression as their disease progresses from the mild to more moderate or severe
stages of the disease. As with those younger patients unaffected by MNCD, the spectrum
of “normal” sexual behavior is wide and undoubtedly influenced by a patient’s under-
lying personality or past sexual experiences. Those with advanced MNCD may even
confuse those they encounter in day-to-day life with a spouse or significant other that has
passed away.
Sexual expression in those with MNCD therefore should not be universally discour-
aged, shamed, or medically treated as an aberrant behavior. Sexual expression such as
consensual physical touch, holding hands, and kissing can be essential to mental health
for those with MNCD who have the desire.
The workup of SIB in long-term care residents starts with a good history, often
obtained from caregivers. It is important to assess the context of SIB by answering
questions such as (1) what exactly is the behavior being demonstrated? (2) what is the
context of the behavior? (3) how often is the behavior occurring? (4) who is being
impacted by the behavior? (5) is the sexual behavior greatly out of proportion to other
disinhibited behaviors? (Figure 36.1).
Pharmacologic management strategies of SIB in those with MNCD are not predict-
able as there are no randomized controlled trials to establish the efficacy or safety of
various medication interventions. A good general approach to management involves first
addressing modifiable environmental factors that could be leading to SIB. Secondly,
medications known to possibly increase changes in SIB should be identified and removed
if possible (benzodiazepines, dopamine agonists). Finally, pharmacologic treatments
specific to the SIB can be trialed. Antidepressants such as SSRIs, trazodone, and
mirtazapine have been used with some success and are generally well tolerated.
Antiandrogens such as medroxyprogesterone, estrogen, and finasteride can be used in
men and have shown benefit. Anticonvulsants such as gabapentin and tegretol have been
used for SIB (Figure 36.2).
In the case presented here, the patient’s behavior was considered problematic as her
sexual behavior was in common areas among other staff and residents. Other residents
were uncomfortable. The dignity of the patient could not be ensured with sexual
behaviors occurring in public places.
Of interest to the psychiatrist was the fact that these behaviors seemed to escalate
in the setting of Mrs. M’s increasing outward signs of depression and anxiety.
Although studies specific to older adult patients are lacking, it is known that anxiety
and depression have been linked to hypersexual behavior. Also, anxiety and depres-
sion are the most common mental health diagnoses in those who could be considered
to have dysfunctional sexual behaviors. It was reasoned that the patient’s dysfunc-
tional sexual expression could be one of her manifestations of depression or anxiety.
This was the impetus to initiate more aggressive treatment with an SNRI/atypical
antipsychotic combination. Staff were encouraged to continue attempts to redirect
Mrs. M to private areas of the facility where she could express herself without
offending others and while maintaining her dignity. It was hoped that as the treatment
of her potential underlying mood disorder took effect, she would be less angry and
upset with attempts at redirection.
“Can I Have a Hug?” 187
Take-Home Points
Sexually inappropriate behavior can be a manifestation of disinhibited behavior seen in older adults
with MNCD.
The evaluation of SIB should include a detailed history of what the behavior is, who is involved,
what the context of the behavior is, who is being impacted, and if the behavior is out of proportion
with other disinhibited behaviors.
Sexual expression in older adults with MNCD is normal and should not be universally discouraged
when in the appropriate setting and when the rights of others are not violated.
Look for and potentially remove medications that could be contributing to SIB including dopamine
agonists, anticholinergic agents, and benzodiazepines.
37 Pseudobulbar Affect
Mr. P was a 78-year-old who had been a resident of a local long-term care facility since
the death of his wife several years earlier. He had a history of coronary artery disease,
hypertension, mild obesity, diet-controlled diabetes, benign prostatic hyperplasia, and a
stroke. Before his stroke, Mr. P was quite functional and independent. The stroke left
Mr. P with left-sided weakness but after extensive rehab he was able to ambulate with the
assistance of a walker. He was also left with some mild cognitive impairment. He could
generally complete his own ADLs with reminders from his wife or other caregivers.
Sadly, after the death of his wife from breast cancer the patient was forced to enter a
long-term care facility as his children did not live close and were very active in their own
careers and with their own families. Mr. P did surprisingly well with the transition to
long-term care. He made friends at the facility, enjoyed participating in social activities,
and got along well with others. He adapted well to the day-to-day routine of his facility.
The geriatric psychiatry team was consulted for the evaluation of potential depression.
Mr. P was noted to become very emotional and have episodes of crying at times, which were
usually short-lived but disturbing to the patient and those around him. His primary
physician had placed Mr. P on escitalopram 5 mg daily and this was subsequently increased
to 10 mg daily. He had been on the 10 mg daily dose for approximately six weeks prior to
psychiatry being consulted but there was little improvement noted in his emotional lability.
Mr. P was on an additional medication regimen of low-dose aspirin, atorvastatin,
metformin, lisinopril, multivitamin, and tamsulosin. He did not have any prior signifi-
cant psychiatric history.
When interviewed Mr. P was friendly and pleasant. He scored 22/30 on the SLUMS
exam and scored 3 on the PHQ–9. He had left-sided weakness and some mild dysarthria
consistent with his history of stroke. His mood was described as euthymic and his
thought processes were thought to be logical. He had no evidence of any hallucinations
or delusions. He was noted to become suddenly tearful when the conversation came
around to discussing one or two of the caregivers at the facility when he mentioned how
grateful he was for the day-to-day help and care they provided him with. He was also
quite tearful when offering thanks to the consulting psychiatrist for the attention
provided to him. “Everyone is just so good to me!” he sobbed. When asked, he felt that
the escitalopram started six weeks before had maybe been a little helpful with his
episodes of crying.
After reviewing the patient’s medical records and speaking to caregivers a presump-
tive diagnosis of MNCD–vascular type–mild with secondary pseudobulbar affect (PBA)
was made. The patient scored 14 on the Center for Neurologic Study – Lability Scale
(CNS–LS scale). This score was suggestive of pseudobulbar affect.
188
“Everyone Is Just So Good to Me!” 189
Teaching Points
Pseudobulbar affect is described as episodes of involuntary, unprovoked, and sudden
episodes of crying or laughing that happen as a result of CNS pathology. Pseudobulbar
affect can be easily overlooked or misdiagnosed as a primary mood disorder. Over the
years it has been called a number of different names including emotional incontinence,
pathological laughter and crying, and excessive emotionalism, among others.
Pseudobulbar affect is seen in a wide variety of neuropsychiatric disorders including
Parkinson’s disease, ALS, Alzheimer’s disease, multiple sclerosis, history of stroke, TBI,
and CNS malignancies. The burden of this disorder can be high, causing embarrassment
and distressing impairment in social situations. The displays of emotion cannot be
prevented or aborted by the patient, leading to social isolation and contributing to
anxiety and depression. Symptoms can include emotionally congruent reactions that
are pathologically intense. Patients can also demonstrate emotionally incongruent affect
reactions such as crying during a happy occasion or while watching a TV sitcom.
The exact pathophysiology behind the disorder is poorly understood but thought to
involve several pathways in the brain that control motor, cognitive, and affective
function. Pathological crying seems to be more common than pathological laughter.
There can be a good deal of overlap in the symptomatology of patients with PBA and
primary mood disorders. Many of the primary neurological disorders associated with
PBA also have a high incidence of comorbid anxiety and depression. When trying to
distinguish anxiety and depression from PBA it should be noted that symptoms of
depression and anxiety tend to be more persistent throughout the day, whereas PBA
symptomatology tends to be more sporadic.
There are four major diagnostic criteria for PBA. These include the presence of an
emotional response that is situationally inappropriate, the patient’s feelings and the
effective response not being closely related, the patient being unable to control the duration
and severity of the episodes, and expressions of emotion not leading to a feeling of relief.
Diagnosis is usually made by an appropriate history with congruent symptomatol-
ogy, but rating scores do exist that are helpful. A common one is the CNS–LS. It is a
seven-item questionnaire that can be self-administered or administered by a healthcare
professional.
Several classes of medications have been used with varying success [1] (see Box 37.1).
Treatments generally focus on the neuromodulation of norepinephrine, serotonin, or
glutamate. Most treatments have been used off-label until dextromethorphan/quinidine
(Nuedexta) was approved in 2010. Dextromethorphan acts as a noncompetitive NMDA
receptor antagonist and is rapidly and extensively converted in the liver to a compound
that is unable to cross the blood–brain barrier. Quinidine helps to slow this hepatic
190 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
Take-Home Points
38 Dementia?”
ADD in Older Adults
Mr. A was a boisterous 78-year-old gentleman who resided at a long-term care facility.
He was quite popular with other residents and staff alike as he was very sociable and
friendly and enjoyed participating in many of the facility’s activities. He moved into the
long-term care facility two years prior after a fall at home which resulted in a hip
fracture. Before this, he was living in a small travel trailer on a piece of land owned by
a friend. He had never been married and had no children.
During hospitalization after the fall, a psychiatry consultation was ordered after the
care team questioned his ability to live alone due to possible dementia. He was evaluated
by a psychiatric nurse practitioner who diagnosed the patient with mild to moderate
MNCD, likely Alzheimer’s type. Mr. A’s SLUMS score was 18/30 at that time. After input
from the mental health practitioner, the medical team recommended the patient be
transferred to an acute rehab facility and likely to long-term care afterward due to the
inability to care for himself secondary to dementia.
Mr. A had several medical problems that contributed to his physical decline, includ-
ing poorly controlled diabetes, COPD, CHF, and mild renal disease. All of these condi-
tions became well controlled as he was having his medications reliably administered by
facility staff and getting regular medical checkups. In the long-term care facility, he was
able to perform his basic ADLs without problem. Despite the diagnosis of probable
Alzheimer’s disease, Mr. A had no apparent declines in either cognition or independence
in the two years he had been at the facility.
Over the course of several months, Mr. A’s long-term care PCP observed him to
be quite cheerful and socially appropriate. He knew the names of nearly all the other
residents. He liked to play bingo, loved to eat, and slept well. His only visitor from
outside the facility was his friend who owned the land where he used to live. During
one of the visits the provider met the patient’s friend and he and Mr. A reminisced.
Mr. A had lived on the piece of property in his travel trailer for 20 years. He had
never worked a “real job” but always seemed able to support himself with the help
of food stamps and odd jobs, mostly consisting of small engine repairs for his
friend who owned an equipment rental shop. His front yard was well known to be
piled high with various machines and small engine parts, most rusted beyond
recognition.
Mr. A had never graduated high school. He had made it through the eighth grade but
dropped out of his small country school at the age of 13. He mentioned that he had been
a terrible student but had always been good at “figuring out how things work”. There was
a time in his life when Mr. A admitted that he abused alcohol. He lost his driver’s license
due to several DUIs in his 30s and it had never been reinstated. He quit drinking
191
192 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
altogether in his 40s after he had a bad fall at home and hit his head, suffering a bad
concussion. He did not smoke or have any history of using recreational drugs.
His parents had both died when he was relatively young and he had no siblings.
He had come close to getting married on a couple of occasions but says, “I saw the light
before it was too late!”
Mr. A said his hobby had always been fixing things. He did not particularly enjoy
television, except for game shows and a few sitcoms, as he says he had a hard time
following along during a movie or an hour-long drama-type show.
Mr. A asked, “Why do they say I have dementia?” He saw people around the facility
with dementia and did not feel that he had similar issues. Another SLUMS exam was
performed and the patient scored 18/30. It was noted that Mr. A had a very difficult time
staying focused on the SLUMS as it was being administered, taking about twice as long as
it does for most other people.
The provider began to suspect that Mr. A’s cognitive difficulties were due to inatten-
tion and possible adult attention deficit disorder (ADD). With further questioning,
Mr. A revealed that he did quite poorly in school when he was young. He relayed that
he had hated school, always felt picked on by the teachers, and his desk was always placed
in the front corner of the room away from the other children for speaking out of turn.
He was always getting in trouble for daydreaming and never turning in his homework.
At one point his parents were told that Mr. A had a learning disability but Mr. A says
“they didn’t know what to do about it back then”.
Mr. A was informed by his provider about the likelihood that he had ADD and how
this diagnosis could sometimes cause symptoms that can be confused with dementia in
older adults or learning disabilities and behavioral problems in children. Mr. A was not
inclined to consider any treatment for the condition as he felt he was doing well and felt
well-adjusted to his new life and community. He also stated, “I already take too many
pills!”
Teaching Points
Attention deficit hyperactivity disorder (ADHD) was for many years considered to be a
condition primarily affecting children and adolescents. We now know that up to 60% of
children and adolescents retain symptoms of ADD into adulthood [1]. Symptoms such
as difficulty concentrating, forgetfulness, and difficulty with organization or executive
function can mimic cognitive issues seen in those with early MNCDs. This distinction
can be particularly difficult in those older adults who present with cognitive complaints.
Approximately 5–7% of children and adolescents are thought to have ADHD. The
condition also affects an estimated 2.5% of adults across the world [2]. The condition is
more commonly diagnosed in males than females with a male-to-female ratio of about
2:1. The effects of ADHD in adults and older adults can involve multiple areas of life
including professional activities, social activities, financial health, and relationships.
In this age of heightened awareness of ADHD, it has now become less likely for a patient
to present to a healthcare provider with a many-year history of undiagnosed or
untreated ADD.
The pathophysiology of ADD is complex and not completely understood but, as with
many neuropsychiatric conditions, it is believed to involve a combination of genetic,
neurobiological, and environmental factors. One leading theory involves the dysregulation
“Why Do They Say I Have Dementia?” 193
Specify whether:
F90.2 Combined presentation: If both Criterion 1a (inattention) and Criterion 1b
(hyperactivity–impulsivity) are met for the past six months.
F90.0 Predominantly inattentive presentation: If Criterion 1a (inattention) is met but
Criterion 1b (hyperactivity–impulsivity) is not met for the past six months.
F90.1 Predominantly hyperactive/impulsive presentation: If Criterion 1b
(hyperactivity–impulsivity) is met and Criterion 1a (inattention) is not met for the past
six months.
Specify if: In partial remission: When full criteria were previously met, fewer than the full
criteria have been met for the past six months, and the symptoms still result in impairment
in social, academic, or occupational functioning.
Specify current severity:
Mild: Few, if any, symptoms in excess of those required to make the diagnosis are
present, and symptoms result in no more than minor impairments in social or
occupational functioning.
Moderate: Symptoms or functional impairment between “mild” and “severe”
are present.
Severe: Many symptoms in excess of those required to make the diagnosis, or several
symptoms that are particularly severe, are present, or the symptoms result in marked
impairment in social or occupational functioning.
The prognosis for adult ADHD is generally good with appropriate treatment and
follow-up. These patients can function well in professional, social, and relationship
arenas. Older adults may potentially have less need for treatment as their symptoms
may be less disruptive to day-to-day function.
196 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
Take-Home Points
39 Any Sleep!”
Sleep–Wake Cycle Disorders
Mr. R was an 82-year-old gentleman who had resided in a long-term care facility for
three months. He possessed an outgoing personality and was generally a favorite among
the facility care staff and other residents. He had a history of mild MNCD, likely
Alzheimer’s type, mild COPD, psoriasis, and osteoarthritis involving multiple joints.
Before moving to the long-term care facility he had lived with his daughter, who had
experienced a decline in her own health necessitating his move to long-term care.
Mr. R was quite content with his move to the long-term care facility, actively
participating in activities and enjoying communal dining. He mentioned that living with
his daughter had often left him feeling bored and, at times, like a burden. He was
considered eccentric in one or two ways, such as his preference to have his evening meal
as early as possible and often being found in bed asleep for the night around 6 or
6:30 pm. Despite sleeping soundly, he would frequently wake up around 2 am and could
be found sitting at the nurse’s station, engaging in conversations with the night shift staff
and watching old westerns on the television in the common area.
About a month before a psychiatric consultation was requested, Mr. R acquired a
roommate, allowing him to pay a significantly lower monthly fee for his room and board
at the facility. Several weeks after getting the new roommate, Mr. R was reportedly
irritable and less pleasant to be around. The facility staff began to wonder if Mr. R was
becoming depressed because of the dramatic change in his personality, leading to the
request for a consultation with the long-term care psychiatrist.
During the initial visit, Mr. R was observed in his room at 2 pm in the afternoon,
nodding off to sleep in his wheelchair. While he was cooperative with the interviewer, he
was best described as irritated and appeared tired. He was reasonably well-groomed and
appeared well-nourished. He was oriented to person and place and able to answer all
direct, in-the-moment questions but struggled as a historian and exhibited some forget-
fulness. His mood was described as irritable. When asked about his feelings toward the
day-to-day routine at the facility and the staff, he reported having no complaints, except
for his roommate. Under his breath, he expressed, “I hate that guy . . . I can’t get any
sleep! He’s up till one or two in the morning watching Fox News and then sleeps all
morning, expecting me to be quiet so he can sleep till noon!”
Mr. R was not taking any psychiatric medications. He denied feelings of sadness or
depression but admitted to irritability and fatigue. He also denied experiencing any
hallucinations. When questioned about his early bedtime, he explained that during his
35 years working in construction, he had to leave his house at 4 am in the morning,
leading him to become accustomed to going to bed early and waking up very early.
197
198 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
Mr. R mentioned to the provider that his new roommate had worked the overnight shift
at a large brewery for 30 years.
The psychiatrist found no evidence of mood disorder, anxiety disorder, thought
disorder, or rapidly progressing MNCD. Mr. R was informed that his circadian rhythm
or “internal clock” was now set for sleep times much earlier than most people, likely due
to his years of working early mornings in construction. This pattern, known as advanced
sleep phase syndrome, is not inherently problematic unless it significantly impairs day-
to-day functioning. Ironically, his new roommate likely had delayed sleep phase syn-
drome due to years of working nights. The suggested options to address this issue were to
either change roommates or, for both Mr. R and his roommate, to gradually adjust their
sleep cycles to a more conventional schedule.
Mr. R and his roommate both expressed a desire to make their current living
situation work and were open to efforts to bring about change. Mr. R agreed to delay
his bedtime by half an hour every week until he reached a more conventional schedule.
Simultaneously, his roommate agreed to advance his bedtime by half an hour per week
until reaching a more conventional time.
During a follow-up visit six weeks later, both men were doing well. Mr. R was now
going to bed at around 9:30 pm at night, spending some time watching Fox News with
his roommate before they retired to bed at approximately 10 pm. Both men had
gradually adjusted to their new sleep schedules and were developing a friendship in
the process.
Teaching Points
Sleep-related complaints are quite common in the long-term care setting. Some may be
symptoms of psychiatric disorders, but many can be attributed to primary sleep dis-
orders. It is estimated that 40–70% of older adults experience some form of chronic
sleep-related complaints, and up to 50% of these cases go undiagnosed [1].
The physiology of sleep in older adults is well known to exhibit differences when
compared to that of younger adults. Box 39.1 lists some age-related changes to sleep
physiology [2,3].
Poor sleep quality and fatigue can result from other physiological or psychiatric
conditions, but they can also be primary issues related to circadian rhythm disturbances.
Circadian rhythms encompass physical, mental, and behavioral patterns that follow a
Box 39.1 Some age-related changes to sleep physiology seen in older adults
Increased:
Time awake after sleep onset (wakefulness at night).
Number of sleep arousals.
Sleep latency-time to fall asleep.
Decreased:
Total sleep time.
Slow wave sleep.
REM sleep.
Sleep efficiency.
“I Hate That Guy. . . I Can’t Get Any Sleep!” 199
24-hour cycle. The most evident example of a circadian rhythm is the sleep–wake cycle,
which typically involves sleep during the night and wakefulness during the day. Various
other bodily systems are closely linked to circadian rhythms as well, including endocri-
nologic hormone release, eating habits and digestion, urine production, and body
temperature, among others.
The neurons of the suprachiasmatic nucleus (SCN) of the anterior hypothalamus are
the heart of the circadian rhythm in mammals. They influence other parts of the brain
via efferent neurons and the humoral effects of melatonin released from the pineal gland.
The activity of the SCN runs autonomously but is regulated by environmental signals.
It resets daily and is heavily influenced by light inputs from the retina during the day and
melatonin secretion at night [4].
Significant distress and dysfunction can arise when there is a misalignment between
the endogenous biological clock and the external light–dark cycle. External factors
influencing circadian rhythms can include light exposure, work schedules, eating sched-
ules, and exercise.
Disturbances in the sleep–wake cycle are also referred to as circadian rhythm
disorders. Box 39.2 lists some of the more common sleep–wake cycle disturbances.
The treatment of sleep disorders in older adults depends on the primary diagnosis.
Long-term care physicians are often the ones tasked with making these diagnoses, as access
to a sleep medicine specialist may be challenging for long-term care residents. Some
general principles to encourage higher-quality sleep in older adults are listed in Box 39.3.
Box 39.2 Some common sleep–wake cycle disturbances seen in older adults
Insomnia: These patients have trouble initiating sleep, often lying awake for an extended
period before falling asleep. Insomnia also includes those who have trouble with
maintaining sleep, who may have frequent awakenings during the night and difficulty
returning to sleep.
Delayed sleep phase syndrome: These patients tend to go to bed and wake up later than
what is considered normal. This can lead to difficulties in day-to-day life and
social function.
Advanced sleep phase syndrome: This is considered the opposite of delayed sleep phase
syndrome. These patients tend to go to bed and wake up earlier than is conventionally
considered normal.
Irregular sleep–wake rhythm: This sleep disorder can lead to multiple short sleep
episodes throughout a 24-hour day. There may be no clear pattern of consolidated sleep
at night.
Non-24-hour sleep–wake disorder: These patients may have longer than 24-hour
circadian rhythms, causing their sleep–wake pattern to be later and later each day,
making it difficult to maintain a schedule.
Sleep state misperception (paradoxical insomnia): These patients consistently
underestimate the amount of sleep they get. They may complain of insomnia, but
objective measurements of sleep duration and quality are normal.
Hypersomnia: Patients with hypersomnia have excessive daytime sleepiness that can be
due to poor quality sleep, sleep disorders, or underlying medical or
psychiatric conditions.
Narcolepsy: A relatively rare neurological disorder that leads to excessive daytime
sleepiness, sudden loss of muscle tone, and other REM sleep-related symptoms.
200 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
Box 39.3 Good nonpharmacologic principles beneficial to sleep quality in older adults
Sleep hygiene practices: These may include maintaining a consistent sleep schedule,
creating a comfortable and dark sleeping environment, avoiding stimulating activities
and bright screens before bedtime, and limiting caffeine intake in the latter part of
the day.
Sleep restriction: Limiting time spent in bed to improve the quality of sleep while in bed.
Physical activity: Physical activity during the day can promote better sleep at night. Limit
physical activity around bedtime.
Limiting naps: Daytime napping can reduce sleep quality at night.
Light exposure: Bright natural light exposure during the day can help maintain circadian
rhythms and better sleep quality at night.
Take-Home Points
Sleep-related complaints are quite common in the long-term care setting. It is estimated that
40–70% of older adults have some type of chronic sleep-related complaint. Up to 50% of these
go undiagnosed.
Older adults are known to have increased wakefulness at night, increased number of sleep arousals,
and increased sleep latency, alongside decreased total sleep time, slow wave sleep, REM sleep, and
sleep efficiency.
Some common sleep–wake cycle disturbances seen in older adults include primary insomnia,
delayed sleep phase syndrome, advanced sleep phase syndrome, irregular sleep–wake rhythm,
non-24-hour sleep–wake disorder, sleep state misperception, hypersomnia, and narcolepsy.
Good nonpharmacologic principles beneficial to sleep quality are often safer and more effective
than pharmacologic therapies. These include good sleep hygiene practices, sleep restriction,
increased physical activity, limiting daytime naps, and daytime bright light exposure.
“I Hate That Guy. . . I Can’t Get Any Sleep!” 201
Ms. H was a 78-year-old woman who had been living in a nursing home for two years.
Since she was admitted, staff had noticed that Ms. H had tried to invite various male
residents to her room and had tried to kiss them or would ask for “just one kiss.”
A consultant psychiatrist was requested because of the staff’s report that Ms. H was
“sexually aggressive” toward male residents.
After a detailed interview and input from various staff members, the psychiatrist
discovered that the nursing assistants who were involved in caring for Ms. H for the first
year and a half had left. The new nursing assistants had just obtained their certifications
and were unsure how to deal with Ms. H’s SIB. One nursing assistant felt that the
behavior was “cute” and “funny” and even giggled when Ms. H exhibited SIB, while
another felt the behavior was “abhorrent” and needed to be stopped. In fact, the latter
nursing assistant had told Ms. H that she was not behaving like a “lady” and another staff
member found Ms. H crying after this exchange.
The “sexually aggressive” behavior that precipitated the consult was that Ms. H had
succeeded in getting a male resident into her room and they were found kissing. During
the interview, Ms. H was a cheerful woman who asked the psychiatrist to give her “just
one kiss.” The psychiatrist showed her his wedding ring, but Ms. H responded by stating,
“Your wife will not know.” The psychiatrist told her that he loved his wife and could not
grant Ms. H’s wish. The clinician was pleasant and nonjudgmental during the entire
interview and shook Ms. H’s hand at the end. The psychiatrist spent a lot of time
educating the staff caring for Ms. H that her need for sexual gratification was normal,
although expressed inappropriately because of her MNCD, and emphasized the import-
ance of being nonjudgmental and the need for a consistent staff approach. The psych-
iatrist also discussed the role of damage to the frontal lobe or its connections in
predisposing the resident to sexually disinhibited behavior.
Over the next eight weeks, Ms. H responded well to a nonjudgmental approach and
firm and consistent redirection by all staff. Ms. H was encouraged to hug and hold hands
with male residents who liked it (and whose family agreed with such contact) and her SIB
became much more manageable. The psychiatrist also met with the director of nursing
and administrator and together they decided to have facility-wide in-service training for
all staff addressing this topic.
Teaching Points
A significant percentage of older adults remain sexually active [1]. Studies have shown
that sexual activity in older adults is associated with better cognitive and mental health
202
“Just One Kiss” 203
and better sleep. Quality of life and general satisfaction may also be adversely affected by
the decline in sexual activity with age [2].
The development of MNCDs in patients does not preclude the need for a sexual life
but does alter the way love is given and received. The misconception that those with
cognitive impairment are asexual is commonly used by caregivers to discourage them
from seeking fulfillment of their sexual needs.
Major neurocognitive disorders are a common cause of SIB. Despite the few studies
discussing sexual aspects in MNCD, existing reports indicate that up to 25% of patients
suffering from Alzheimer’s disease have inappropriate sexual behavior, including
increased sexual urge and hypersexuality [3,4]. These behaviors can result in increased
burden on caregivers and clinicians, increased use of psychotropic medications,
increased utilization of healthcare resources, and earlier transition to long-term care
facilities [3].
Neurobiological changes in MNCDs may result in a stronger impulse to have sex,
decreased sex drive, sexual aggression, anger in response to rejection, or even a complete
loss of interest in sexual expression. During the early stages of Alzheimer’s disease
specifically, the sexual desire frequently increases or, alternatively, there is a complete
loss of desire for sexual expression [5].
Dysfunction of various anatomical areas of the brain has been implicated in the
neurobiology of inappropriate sexual behavior. Anatomic areas of particular importance
in SIB include the frontal lobe, the temporal limbic system, the striatum, and the
hypothalamus. Frontal lobe pathology can lead to more disinhibited behaviors such as
public exposure or public masturbation. Degeneration of the striatum can be more
associated with OCD sexual behaviors. Pathology of the temporal limbic system can
lead to increased sexual drive as seen in Kluver–Bucy syndrome. Lesions in the hypo-
thalamus can also be seen in those with hypersexuality [6].
Definitions of SIB in patients with MNCD can vary widely depending on who is
asked the question. Definite acts of SIB include making explicit sexual comments,
touching someone other than a partner on the breasts or genitals, touching a partner
on the breasts or genitals in public, requesting unneeded genital care, and exposing the
genitals or breasts in public. More ambiguous demonstrations of SIB can involve being
undressed outside of the bedroom or bathroom, seeking out non-genital intimate touch
such as kissing or caressing another, or self-touch of genitals through one’s clothes [7].
Since there is no widely agreed-upon definition of SIB the distinction is usually based on
the judgment of what is normal for a person in a particular situation or on the level of
risk or discomfort of others.
Nursing assistants at long-term care facilities are responsible for the brunt of the day-
to-day patient care. Despite their importance, they likely receive little instruction on
issues relating to mental health in those with MNCD, especially sexuality. Turnover rates
are also quite high in many long-term care facilities, which makes education even more
challenging. Nursing assistants and other care staff should be made aware via educational
opportunities that sexual behavior can increase in those with MNCD due to the
following reasons: (1) there is brain dysfunction due to the underlying disease process,
(2) there can be disease-related disinhibition of sexual behaviors, (3) normal etiquette
can be forgotten in illnesses with cognitive impairment, (4) delusions and hallucinations
may impact sexual behavior. Staff should also be informed about social and environ-
mental factors that can play a role in increased SIB. Examples of this include the loss of a
204 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
usual sexual partner, lack of privacy, an under stimulating environment, and loss of the
ability to properly interpret social cues. Staff should also be made aware that certain
medications have the potential to increase SIB. Understanding the reasons for SIB can be
helpful for those who must deal with these behaviors on a day-to-day basis [7].
There are instances when the sexual expression of long-term care residents should be
particularly protected and encouraged. It is not uncommon for a married or partnered
couple to room together in an AL or long-term care setting. One or both of the couple
can have cognitive challenges. In these situations, it may become important to assess a
person’s capacity to consent to sexual activity and to possibly reassess capacity should
cognition continue to decline [8].
The ability of patients to consent to sexual relations may be underestimated by
healthcare staff. Therefore, the issue of capacity to consent to sexual activity is an
important point of discussion among staff members and each case should be assessed
separately. Some experts recommend assessment of these situations by a geriatrician, an
internist experienced in the care of older adults, or a geriatric psychiatrist [9]. The need
for expert opinion supports the notion that assessing a patient’s capacity to consent to
sexual activity can be difficult. Questions pertinent to assessing a patient’s ability to
consent to sexual activity are shown in Box 40.1.
Take-Home Points
A significant percentage of older adults remain sexually active. Studies have shown that sexual
activity in older adults is associated with better cognitive and mental health and better sleep.
Major neurocognitive disorders are a common cause of SIB. Despite the few studies discussing the
sexual aspects of MNCD, existing reports indicate that up to 25% of patients suffering from
Alzheimer’s disease have inappropriate sexual behavior, including increased sexual urge
and hypersexuality.
The ability of patients to consent to sexual relations may be underestimated by healthcare staff.
Therefore, the issue of consent is an important point of discussion among staff members and each
case should be assessed separately.
Long-term care facilities should have policies in place that address the evaluation of a patient’s
capacity to consent to sexual activity.
“Just One Kiss” 205
41 Anticholinergic Medications,
Incontinence, and Overactive Bladder
Mrs. S was a 72-year-old woman who had multiple chronic medical problems (diabetes
for 30 years, hypertension, osteoarthritis, chronic liver disease due to alcoholism,
peripheral neuropathy) and recently had right-sided hemiplegia due to stroke. After
some initial anxiety and depression upon moving into an AL home, she had been doing
well for three months. Mrs. S was a frail, petite woman who had a body mass index of 17.
She was taking 12 different medications for her many medical conditions, including
10 mg of amitriptyline for peripheral neuropathic pain and insomnia. For several weeks,
Mrs. S had developed urinary frequency and incontinence and was started on oxybutynin
5 mg daily for suspected overactive bladder (OAB). Her urine analysis was negative
for infection.
The staff noticed that over five days the resident had become more confused and
irritable, had two falls (without injury), and was unable to sleep more than two or three
hours at night. The provider recommended discontinuing amitriptyline, but the resident
and family refused because she had been taking it for more than 15 years and it had helped
her peripheral neuropathy pain and insomnia. A psychiatric consult was obtained.
Mrs. S told the psychiatrist, “I don’t feel well,” but could not clarify what was
bothering her. After the assessment, the psychiatrist recommended discontinuing oxy-
butynin and following a scheduled toileting program to treat OAB. Over the next
72 hours Mrs. S’s delirium resolved, and she was back to her level of functioning before
starting oxybutynin.
The psychiatrist then met with the resident and her family, explaining the risk of
cognitive impairment with amitriptyline and the potential benefits of alternatives, such
as gabapentin or nortriptyline. The family agreed to try switching to gabapentin with the
assurance that if it did not help, the resident could restart the amitriptyline. The
psychiatrist checked the resident’s renal function before she started taking the gabapen-
tin. Her serum creatinine was 1.8, indicating mild chronic kidney disease. Amitriptyline
was decreased to 5 mg daily at bedtime for two weeks and then discontinued. Gabapentin
was added at 100 mg daily at bedtime and over two weeks increased to 300 mg at
bedtime. The resident had mild insomnia for a few days but no increase in peripheral
neuropathy pain. To improve sleep, the staff was recommended to encourage Mrs. S to
increase physical activity, increase exposure to sunlight, and minimize daytime napping.
After two weeks of taking gabapentin, discontinuing amitriptyline, and beginning a
regimen of individualized psychosocial–spiritual interventions, Mrs. S felt she was doing
“quite well.” The staff noticed that her constipation was replaced with mild diarrhea,
which resolved once the medications for constipation (docusate sodium and senna)
were discontinued.
206
“I Don’t Feel Well” 207
Teaching Points
Adding an anticholinergic medication such as oxybutynin for a frail resident who is
already taking other medications with high anticholinergic activity (amitriptyline in this
case) and compromised neurological status (cerebrovascular disease in this case) puts
that resident at high risk of drug-induced delirium. Drugs for OAB such as tolterodine
and oxybutynin and other anticholinergic drugs (diphenhydramine, hydroxyzine) may
precipitate delirium and should be used with great caution in residents who are frail,
have pre-existing MNCD, have a history of liver disease, or are taking multiple drugs
metabolized through the Cytochrome PCY450 system [1]. A rigorous trial of scheduled
toileting should be considered before prescribing drugs for OAB.
Each assessment for change in mental status is also an opportunity to review the
resident’s medications and reduce the anticholinergic load. Gabapentin has no antic-
holinergic activity and is a much better choice than amitriptyline for the treatment of
peripheral neuropathy in long-term care residents. Because gabapentin is primarily
excreted by the kidneys, it is important to start with a low dose in residents who have
chronic kidney disease, and a very low dose may be sufficient for a therapeutic response.
Additionally, for a resident who has been taking psychiatric medication for a long time,
discontinuing such medication is a major decision and may be associated with significant
anxiety. Hence, it is important that the healthcare provider discuss the risks of continu-
ing current psychiatric medications and the benefits of safer alternatives before institut-
ing any change. Finally, anticholinergic medications are constipating, and most residents
need medication to treat constipation caused by anticholinergic drugs. By reducing the
anticholinergic load, the healthcare professional may help residents decrease the need for
medication to treat constipation, thus reducing polypharmacy and its harmful
consequences.
The commonly encountered issues of urinary incontinence in the older patient
population are such that it warrants further discussion here. The prevalence of urinary
incontinence in women aged 70 years and older in long-term care settings is estimated to
be around 50–80%. The prevalence of urinary incontinence in men over 70 in long-term
care settings varies depending on the study and population examined but is thought to be
closer to about 45%. Urinary incontinence in older patients can have significant psychi-
atric implications, including depression, anxiety, social isolation, and decreased quality
of life. Incontinence can cause embarrassment, shame, and loss of dignity, leading to
feelings of frustration, helplessness, and hopelessness. Nygaard et al. reported that 80% of
women with severe urinary incontinence are likely to develop depression compared with
40% of those with mild incontinence [2]. Incontinence can also interfere with daily
activities, including work and social engagements, further exacerbating feelings of
isolation and depression. Patients with incontinence may become more reliant on
caregivers and may experience a loss of autonomy, leading to decreased self-esteem
and negative self-image. It is important to address the psychological impact of incontin-
ence in older patients and provide appropriate support and treatment to improve the
overall quality of life.
There are six major categories of urinary incontinence. They include stress incontin-
ence, urge incontinence, mixed incontinence, overflow incontinence, functional incon-
tinence, and total incontinence. Stress incontinence occurs when urine leaks due to
physical activity such as coughing, sneezing, laughing, or exercising. Urge incontinence
208 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
involves a sudden and strong urge to urinate, followed by an involuntary loss of urine.
It may be caused by OAB or other neurological conditions. Mixed incontinence is a
combination of stress and urge incontinence. Overflow incontinence happens when the
bladder does not empty completely and leads to constant dribbling of urine. Functional
incontinence occurs when physical or cognitive impairment makes it difficult for a
person to reach the bathroom in time. Finally, total incontinence refers to the continuous
and total loss of urinary control, which may result from a congenital abnormality
or injury.
The differential diagnosis of urinary incontinence in older adults is broad. Box 41.1
includes a list of potential causes [3].
The workup of incontinence in older adults can be time-consuming, inconvenient,
and costly. The most basic and practical workup includes a complete history, physical
exam, medication review, and urinalysis. Those in long-term care often have significant
logistic barriers to seeing a urologist in an outpatient setting for a more complete workup
that might include urodynamics and cystoscopy. For this reason, it is not uncommon to
see patients affected with incontinence trialed on one of the medications that are
approved for OAB in lieu of a full urologic workup to evaluate for other types
of incontinence.
Overactive bladder and its treatment are often encountered by those treating older
adults in long-term care. The definition of OAB by the International Continence Society
describes it as the presence of urinary urgency, with or without urgency urinary
“I Don’t Feel Well” 209
Take-Home Points
Drugs for OAB such as tolterodine and oxybutynin and other anticholinergic drugs (diphenhydra-
mine, hydroxyzine) may precipitate delirium and should be used with great caution in frail
residents, those with pre-existing MNCD, those with a history of liver disease, or those who are
taking multiple drugs metabolized through the Cytochrome PCY450 system.
Urinary incontinence in older patients can have significant psychiatric implications, including
depression, anxiety, social isolation, and decreased quality of life.
There are six major categories of urinary incontinence. They include stress incontinence, urge
incontinence, mixed incontinence, overflow incontinence, functional incontinence, and
total incontinence.
There are two general classes of medications used to treat OAB. These include antimuscarinic
(anticholinergic) agents such as oxybutynin and tolterodine and beta-3 adrenergic agonists such as
mirabegron and vibegron. The beta-3 adrenergic agents are preferred as they are not anticholi-
nergic and are not included on the Beers list.
“I Like Food”
Case
Mr. W was an 81-year-old resident living in an AL home for two years. He was doing
well, taking 10 mg daily escitalopram for the treatment of MDD-like symptoms and
anxiety symptoms secondary to MNCD due to Alzheimer’s disease. Over two years he
gained 50 pounds, and in the last three months he gained 14 pounds (for a final BMI of
42), primarily due to a sedentary lifestyle and excessive eating during and between meals.
Mr. W had struggled with obesity for most of his life and intermittently went to
Weight Watchers to reduce weight and eat healthier. However, due to Alzheimer’s
disease, he had stopped going to Weight Watchers. A psychiatrist was consulted, as
Mr. W had become irritable, had expressed feeling “upset” about gaining weight, and his
wife felt his depression was coming back and antidepressants should be adjusted. The
psychiatrist found Mr. W to be sociable but upset about his weight gain.
Mr. W told the psychiatrist, “I like food.” Certain other pertinent facts came out while
taking a thorough history from the patient, his family, and staff. Mr. W was often found to
have rather large collections of candy, packaged baked goods, sodas, and salty snacks under
his bed. He was found by staff several times per week in bed, surrounded by crumbs,
empty soda cans, and wrappers. This would usually happen several times per week. On a
couple of occasions, Mr. W had eaten so much that he became ill and vomited. He became
defensive and irritable when staff attempted to discuss these behaviors with him. Mr. W’s
wife admitted that she would regularly bring in goodies for her husband but she felt the
amounts were fairly modest (one or two boxes of snack cakes or bags of chips per week).
The staff mentioned that the patient would also regularly visit the snack area of the facility
and use food found there to add to his under-the-bed collection.
Mrs. W revealed that Mr. W had experienced a complex relationship with food for
most of his adult life and said he had always been “a big man”. When he was younger, he
was well known to consume large quantities of food and to frequent buffet restaurants
alone, eating to the point of being quite uncomfortable. He would later feel guilty for this
and become depressed, irritable, and socially withdrawn. He started his career as an
attorney with great promise, but the stress involved with success and advancing responsi-
bilities would often trigger more dysfunctional eating behavior. Eventually, the patient’s
weight became such that it interfered with his ability to go to work, and he worked more
at home. This hindered his career advancement.
The psychiatrist shared with Mr. W, his wife, and staff that he did not think antidepres-
sants needed to be adjusted. They recommended a program of exercise (five minutes of
strength training and 15 minutes of assisted ambulation daily) and nutritional strategies.
Nutritional strategies involved a portion-controlled diet (smaller portions given during the
three meals), replacing consumption of unhealthy snacks (crackers, candy, ice cream) with
211
212 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
fruits and vegetables Mr. W liked (blueberries, mangoes, carrots, celery with a vegetable
dip), and replacing intake of soda with diet soda and water.
Mr. W’s wife was encouraged to bring in healthier snack options for her husband.
Interventions to help add patient-specific enjoyable activities were helpful as well since
boredom was thought to be a trigger for his dysfunctional eating. Weekly supportive
psychotherapy was initiated to further explore Mr. M’s emotional triggers for eating and
to help him with more productive strategies to deal with them.
Mr. W initially resisted this change in diet but, with the staff’s support and his wife’s
encouragement, he acquiesced. Over the next eight weeks, not only did Mr. W lose four
pounds, he also seemed to have more energy, started getting out of the chair on his own
(previously he needed one-person assistance to do this), and began walking around the
facility. Staff also noticed that Mr. W seemed to carry on longer conversations and
showed less irritability.
Teaching Points
For some residents who are obese, simple nutritional changes with a modest increase in
physical activity and lots of support and encouragement can lead not only to modest but
significant weight loss and also improvement in mood and function.
Eating disorders are complex chronic medical conditions that have both psycho-
logical and medical consequences. They are characterized by abnormal eating behaviors
that can lead to significant morbidity and even mortality. Well-characterized eating
disorders that are described in the DSM–5–TR include anorexia nervosa, binge eating
disorder, and restrictive/avoidant food intake disorder, among others. Eating disorders
are more commonly thought of as affecting younger adults but they can also occur or
continue in older adults [1].
The case here depicts a patient with binge eating disorder. The DSM–5–TR diagnos-
tic criteria for binge eating disorder are presented in Box 42.1[2].
There is relatively little research focusing on eating disorders in older adults and they
may be easily overlooked. Stigmas associated with eating disorders and age-related
misconceptions might contribute to underreporting and underdiagnosis of eating dis-
orders in older adults. Studies have estimated that between 1.8 and 3.8% of women over
the age of 60 have indications of an eating disorder [1]. It is known that the most
common eating disorder in older adults is anorexia nervosa. Binge eating disorder is less
common, being reported by 0.6–1.8% of women and 0.3–0.7% of men [3].
The pathophysiology of binge eating disorder involves both biological and psycho-
logical factors. Just as in substance use disorders, dysregulation in neurocircuitry related
to reward and emotional processing might contribute to the development and mainten-
ance of binge eating behavior. Neurotransmitters such as dopamine, which are involved
in pleasure and reward, might be implicated in the compulsive nature of binge eating.
Neuroimaging studies have shown hyperactivity of the medial orbitofrontal cortex and
hypoactivity in the prefrontal network in individuals with binge eating disorder. One
theory suggests that people exhibiting elevated impulsiveness and heightened responsive-
ness to rewards can develop addictive-like reactions to specific foods, such as those rich
in sugar and fats. This can contribute to and maintain binge eating behaviors [4].
Diagnosing binge eating disorder is largely through a comprehensive history.
Clinicians should ask about the age of onset of binge eating episodes, frequency of
“I Like Food” 213
Box 42.1 DSM–5–TR criteria for the diagnosis of binge eating disorder
1. Recurrent episodes of binge eating. An episode of binge eating is characterized by
both of the following:
a. Eating, in a discrete period of time (e.g. within any two-hour period), an amount of
food that is definitely larger than what most people would eat in a similar period of
time under similar circumstances.
b. A lack of control when eating during the episode (e.g. a feeling that one cannot
stop eating or control what or how much one is eating).
2. The binge eating episodes are associated with three (or more) of the following:
a. Eating much more rapidly than normal.
b. Eating until feeling uncomfortably full.
c. Eating large amounts of food when not feeling physically hungry.
d. Eating alone because of feeling embarrassed by how much one is eating.
e. Feeling disgusted with oneself, depressed, or very guilty afterward.
Specify if:
In partial remission: After full criteria for binge eating disorder were previously met,
binge eating occurs at an average frequency of less than one episode per week for a
sustained period of time.
In full remission: After full criteria for binge eating disorder were previously met, none of
the criteria have been met for a sustained period of time.
Specify current severity: The minimum level of severity is based on the frequency of
episodes of binge eating. The level of severity may be increased to reflect other
symptoms and the degree of functional disability.
Mild: 1–3 binge eating episodes per week.
Moderate: 4–7 binge eating episodes per week.
Severe: 8–13 binge eating episodes per week.
Extreme: 14 or more binge eating episodes per week.
episodes, amounts of food consumed, emotions associated with the behavior, comorbid
medical problems, potential triggers for the behavior, history of substance abuse, and
family history of binge eating.
Pharmacologic treatment options for binge eating disorder have provided disap-
pointing results. Comorbid mood and anxiety disorders are more common in this
population and treatment of these conditions may produce related improvements in
comorbid eating disorders. Cognitive behavioral therapy and interventions focused on
behavioral weight loss have been shown to be effective in reducing the frequency and
severity of binge eating episodes [5].
In the case here, Mr. W responded well to behavioral-focused techniques and
emotional support, with the family encouraged to provide healthier options for snacks.
Boredom was thought to be a significant trigger for Mr. W’s binge eating so
214 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
individualized interventions were focused on finding frequent and engaging activities for
him to participate in.
The long-term outlook for older adults with binge eating disorder will usually include
periodic setbacks or relapses. Individualized efforts to prevent relapses should focus on
identifying and mitigating triggers of emotional eating. Areas of future research may
involve medications with newer indications for the treatment of obesity such as inject-
able GLP-1 agonists that increase the feeling of satiety, delay gastric emptying, and may
inhibit the pleasurable feedback mechanisms that contribute to dysfunctional behaviors
in those with binge eating disorder [6].
Ethical challenges can present themselves in dealing with those with eating disorders
in the long-term care setting. Patients have as much right to engage in legal, albeit self-
destructive, behaviors as those outside of long-term care. An overly paternalistic
approach to the treatment of those in long-term care can undermine a resident’s right
to autonomy, dignity, and self-determination. A patient such as Mr. W should not be
disallowed access to foods that are central to his dysfunctional eating. A better approach
is to give Mr. W and his family/caregivers the best biopsychosocial support to encourage
him to make better choices for himself.
In past decades it was commonplace for patients to be placed on restrictive diets by
their healthcare providers in the long-term care setting. Very restrictive diets are generally
contraindicated in older adults. These include salt-free diets for those with hypertension or
CHF, no-sugar diets for diabetics, low-fat diets for hyperlipidemia, and very low-protein
diets for those with chronic kidney disease. These restrictive diets are often so unpalatable
as to risk malnutrition. Nutritional goals in long-term care patients should be a well-
balanced diet with a good balance of macro and micronutrients. Also, patients should
generally not be disallowed their favorite foods when in the final stage of life.
Take-Home Points
Eating disorders are complex chronic medical conditions that have both psychological and medical
consequences. They are characterized by abnormal eating behaviors that can lead to significant
morbidity and even mortality.
The pathophysiology of binge eating disorder involves both biological and psychological factors.
Just as in substance use disorders, dysregulation in neurocircuitry related to reward and emotional
processing might contribute to the development and maintenance of binge eating behavior.
Pharmacologic treatment options for binge eating disorder have provided disappointing results.
Comorbid mood and anxiety disorders are more common in this population and treatment of these
conditions may produce related improvements in comorbid eating disorders.
Ethical challenges can present themselves in dealing with those with eating disorders in the long-
term care setting. Patients have as much right to engage in legal, albeit self-destructive, behaviors
as those outside of long-term care.
Mr. T, a resident of a long-term care facility for 12 months, was increasingly depressed
because his wife had reduced the frequency of her visits to once a week and would visit
for only a few minutes. His wife reported that visiting him was too depressing, as he was
not the active and intelligent person she married. Over time, Mr. T lost weight and
stopped going to group activity programs he previously attended.
He was about to be put on an antidepressant when his mood started to improve.
He began eating better, started taking part in activities, and seemed to be genuinely
happy. This was because Mr. T had developed a friendship with a female resident and
they would spend several hours per day together, watching TV or sharing stories. When
asked what he liked about this new friend, Mr. T replied, “I like the way she laughs.”
Mr. T had documented moderately advanced MNCD, mixed type – Alzheimer’s and
vascular. His last SLUMS exam score was 19/30 and was performed three months prior.
His medical comorbidities included mild CHF, well-controlled Type 2 diabetes, and
hypertension. His history of falls and nighttime wandering triggered his admission to
long-term care one year before.
Mrs. T and the patient’s daughter attended a monthly care plan meeting virtually.
The long-term care facility staff at the meeting also included the director of nursing,
administrator, and social worker. Mr. T’s improved mental health was discussed. Both
Mrs. T and the patient’s daughter were clearly shocked and upset when they found out
that Mr. T had taken up a presumed romantic relationship with another resident at the
facility. They were also angered by the fact that this situation was allowed to happen. The
social worker pointed out that, even though Mr. T had dementia and needed support
with several ADLs, he still had the same emotional needs for closeness and intimacy that
he had prior to his admission to the long-term care environment. It was also pointed out
that the relationship between Mr. T and his new romantic interest was mutual and
consensual and that they both had the ability to voice and act on their wishes to continue
in the relationship. The relationship was not clearly intimate physically beyond sitting
closely together and hand-holding but should they express the desire for this the facility
was required to honor their right to privacy and dignity.
At the next care plan meeting Mrs. T and her daughter were present again and inquired
about the relationship between Mr. T and his new companion. The facility staff told his
family that the two appeared to be continuing to enjoy each other’s company and they
could nearly always be found together. The family of Mr. T’s girlfriend was very happy that
she had found companionship and love when she previously seemed sad and lonely.
Mrs. T and her daughter expressed that, after many conversations and time, they
felt more comfortable with the fact that Mr. T had a romantic relationship outside
216
“I Like the Way She Laughs” 217
of his marriage that added comfort and meaning where there was loneliness and
boredom before.
Teaching Points
This case illustrates the power of friendship, attachment, and intimacy in healing
emotional pain. It also illustrates the resiliency of Mr. T (and many other residents) in
the face of overwhelming adversity.
The topic of sexuality and sexual expression within the context of long-term care is
complex. Medical providers, administrators, nursing staff, and social workers should all be
familiar with the basics when it comes to matters of sexuality in the long-term care facility.
It is a misconception that the need for physical intimacy and sexual expression
declines with age. Although normal biological changes that occur with age can lead to
declining libido, the inner drive for physical and emotional connection will often remain
in older adults indefinitely.
The prevalence of loneliness in long-term care is high despite the large number of
interpersonal interactions possible on a given day between patients, their co-residents,
and care staff. Loneliness is a function of both the number and depth of human
relationships. The depth and quality of connection of an older adult with a romantic
partner in the long-term care setting can lead to a huge boost in quality of life.
The rights of residents of long-term care facilities are clearly stated in the Code of
Federal Regulations (CFR) section 483.10. The CFR is a set of regulations that govern
various aspects of healthcare, including nursing homes and long-term care facilities, in
the United States. Some of these rights guaranteed to residents of long-term care facilities
include the right to dignity and respect, the right to make one’s own choices including
the right to refuse medical treatment, the right to privacy and confidentiality, the right to
freedom from abuse and restraints, the right to have visitors of their choice, and the right
to participate in care planning, among others.
Although the CFR is updated every year it does not specifically address matters of
rights to sexual expression in the long-term care setting. However, Appendix PP in the
CFR does and is also updated regularly. This document states, “Residents have the right
to engage in consensual sexual activity. However, any time the facility has reason to
suspect that a resident may not have the capacity to consent to sexual activity, the facility
must take steps to ensure that the resident is protected from abuse. These steps should
include evaluating whether the resident has the capacity to consent to sexual activity.”
The concept of capacity is key to the discussion of sexual activity in older adults in the
long-term care setting. Evaluating a person’s capacity to consent to sexual activity is key
to ensuring the safety of residents from sexual abuse or exploitation. Capacity is not a
blanket determination but is situational. For instance, the capacity to understand and
evaluate the complexities of estate planning necessary to guide one’s financial affairs may
be lacking when the capacity for entering a romantic or sexual relationship remains.
Also, there can be no surrogate decision-maker when it comes to consent for sexual
activity. Universally accepted criteria for the capacity to consent to sexual activity are
lacking and legal criteria vary state by state.
Individual long-term care facilities should have clearly stated policies involving
sexuality that address a resident’s capacity to consent to consensual physical contact of
a sexual nature. Some important questions to be addressed are shown in Box 43.1 [1].
218 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
Take-Home Points
Older adults retain the need for physical and emotional intimacy far into their later years despite
the presence of cognitive impairment.
The capacity to consent to sexual activity is possible for residents who are able to correctly
recognize a potential partner, express interest in sexual activity, communicate needs and wants
either verbally or nonverbally, and communicate a lack of desire for sexual contact.
The involvement of families in discussions of romantic relationships in the long-term care setting
can be challenging but is improved by education and experience on behalf of providers and staff.
Federal regulations regarding long-term care facilities indicate that residents have the right to
engage in consensual sexual activity if they have the capacity to consent.
Further Reading Smith, L., Yang, L., Veronese, N., Soysal, P.,
Stubbs, B., & Jackson, S. E. (2019). Sexual
Esmail, S., & Concannon, B. (2022). activity is associated with greater enjoyment
Approaches to determine and manage of life in older adults. Sexual Medicine,
sexual consent abilities for people with 7 (1), 11–18.
cognitive disabilities: Systematic review.
Interactive Journal of Medical Research, Srinivasan, S., Glover, J., Tampi, R. R., Tampi,
11 (1). https://doi.org/10.2196/28137 D. J., & Sewell, D. D. (2019). Sexuality and
the older adult. Current Psychiatry Reports,
Jackson, S. E., Firth, J., Veronese, N., Stubbs, 21, 1–9.
B., Koyanagi, A., Yang, L., & Smith, L.
Miscellaneous Neuropsychiatric Disorders Affecting
Section 7 Patients in Long-Term Care
“This Is So Hard”
Case
Mr. L, the 90-year-old husband of Mrs. L, an 85-year-old woman who had advanced
MNCD, commented during an interview with Mrs. L’s physician, “My wife is there, but
she is not there. She needs a wheelchair now and her speech is almost gone. She
recognizes me by face at times but calls me ‘Dad’ when she says a few words that I can
make sense of. She is starting to be combative. This is so hard. I find myself getting angry
even though I know that it is her disease that makes her behave this way. Then I get
depressed. This is so unlike her.”
The psychiatrist treating Mrs. L provided emotional support to Mr. L, suggesting that
he see a social worker or therapist who specializes in helping spouses adjust to MNCD in
their loved one. Mr. L was reluctant but agreed after his daughter promised to accom-
pany him on the first visit to the social worker. After several weekly meetings with the
social worker, Mr. L gradually felt less guilty, was able to express his grief and loss, and
learned ways to make his visits with his wife meaningful and even fun at times. He also
started attending spousal support groups run by the local chapter of the Alzheimer’s
Association. Mr. L resumed his regular exercise program at a local health club after the
psychiatrist indicated that, if he keeps himself physically healthy, he would better be able
to support his wife emotionally through her journey with MNCD. Mr. L also became
more mindful of “little” gifts from his wife, such as her breaking into a big smile when
she saw him approach.
Teaching Points
The devastating effects of MNCDs on those afflicted with them are the focus of a major
portion of this book. They experience loss of physical health, loss of mental health, and
sadly a gradual loss of personality and self. We who care for those with MNCDs
understand that care must be provided, not only to the patients suffering from MNCD
but also to those who care for them. Primary caregivers play a pivotal role in the lives of
those with MNCD by providing essential support and assistance. The demands of the
role can lead to significant stress, adversely affecting the caregiver’s physical, psycho-
logical, and financial well-being. Addressing the grief and stress of a family caregiver can
have the added benefit of improving the emotional well-being of a resident in long-
term care.
Caregivers of those with MNCDs may have a multitude of responsibilities. These can
include, but may not be limited to, assisting with ADLs, providing emotional support,
overseeing medication and medical care, and looking after a loved one’s financial
interests, all while balancing their own needs. Caregiving often becomes increasingly
220
“This Is So Hard” 221
demanding as the disease process progresses and caregivers may experience physical
demands related to their role. Earlier in the course of the disease caregivers will often
gradually assume full responsibility for the major activities of the household, including
cooking, shopping and meal planning, financial planning, and managing medical care
and appointments. This can leave a caregiver with less and less time for self-care. Later in
the course of the disease, caregivers may be responsible for providing or overseeing care
involving even basic ADLs such as bathing, feeding, and mobility.
The source of caregiver stress can include the physical and practical demands of
caregiving. The physical demands often lead primary caregivers to seek outside help in
the home or may necessitate a move to a long-term care facility. The emotional and
behavioral manifestations of advancing neurocognitive disorders can be distressing to
caregivers. Those who provide care or care oversight can experience social isolation as
much of their time is spent in the care of their loved ones. This can lead to loneliness,
anxiety, and depression. Caregivers may experience financial stressors related to the care
of their loved one in the form of lost wages due to caregiving responsibilities or paying
for expensive in-home or out-of-home care. Some caregivers may lack sufficient support
from family and friends and the strain of an uncertain future can also weigh heavily on
their minds.
Providing excellent direct medical care and mental healthcare to seniors with
MNCDs is invaluable. However, those who provide medical care to those with
MNCDs should become familiar enough with outside resources to offer guidance to
caregivers. A list of terms applicable to the resources available for advancing care needs
of those with MNCD is presented here.
Senior apartments. Communities generally for those 55 and over or with disabilities
that may have very limited on-site services such as group transportation or a senior
social worker to help with some ADLs. Rent is often subsidized or income-based.
Senior apartments are appropriate for those with no cognitive impairment to mild
cognitive impairment. Cost=$
Independent living. Communities for seniors that provide additional services such
as meals and transportation. Often comes with the option of privately paid ADL
assistance from home health aides. Appropriate for those with no cognitive
impairment to mild or moderate MNCD. Cost=$$
Residential care homes. Facilities that provide care to those with MNCD are more
home-like and tend to have a higher caregiver-to-resident ratio. Usually for those
with moderate to severe MNCD. Cost=$$$
Assisted living. Communities for seniors that provide meals, some transportation,
activities, medication oversight and administration, and usually some licensed
practical nurse or registered nurse care oversight. May provide specialized memory
care units for those with MNCDs. Appropriate for those with no cognitive
impairment to advanced MNCD. Cost: $$$
Continuing care retirement communities. Ascribe to the concept of “age in place”.
The facilities provide care to seniors of all levels of disability from independent to
long-term care. Seniors can move up or down the levels of care as necessary
depending on care needs. Cost=$$$$
Long-term care. Communities for those who need assistance with multiple basic
ADLs. Full medication management is provided. On-site physician and nursing care.
222 Miscellaneous Neuropsychiatric Disorders Affecting Patients in Long-Term Care
Take-Home Points
Health providers of those with MNCD are in a unique position to provide support to primary
caregivers who may be struggling with the many responsibilities of being a primary caregiver.
Be sensitive to the presence of potential caregiver stress. Offer suggestions to caregivers that may
assist in better interactions with their loved one with MNCD.
Become familiar with the various support resources and supported living environments available to
those with MNCD. Having a connection with a social worker or geriatric case manager can be
helpful as a referral source for those families that may need more extensive assistance in planning
for care needs.
Be familiar with and ready to refer family members of those with MNCD to local support groups
such as the Alzheimer’s Association for ongoing caregiver support.
Have a couple of book recommendations on hand that may offer assistance to caregivers such as
The 36-Hour Day: A Family Guide to Caring for People Who Have Alzheimer’s Disease and other
Dementias or What to Do Between the Tears: A Practical Gide to Dealing with a Dementia or
Alzheimer’s Diagnosis in the Family.
residential care homes, 221 take-home points, 96 sleep disorders, See also REM
respiratory complications, 7 teaching points, 95–96 sleep behavior disorder
respiratory depression, 119 sedative-hypnotic (RBD)
respite care, 222 medications, 181 pharmacologic treatment of,
restrictive diets, 214 sedatives, 131 200
retrocollis, 64 seizures, 119 traumatic brain injury, and,
rights of residents of long- selective serotonin reuptake 14
term care facilities, 217 inhibitors, See SSRIs sleep physiology
rigidity, 107 senior apartments, 221 age-related changes, 198
riluzole, 22 sensory deprivation, 3 sleep quality
risperidone serotonin, 79 principles beneficial to, 200
in management of bipolar dysfunction, 86 sleep state misperception, 199
disorder, 90, 91 syndrome, 156 sleep–wake cycle disorders,
in treatment of BPSD, 43 serotonin-norepinephrine 197–198
in treatment of reuptake inhibitors, take-home points, 200
schizoaffective See SNRIs teaching points, 198–200
disorder, 127 sertraline SNRIs
in treatment of in treatment of BPSD, 41 anxiety, and, 154
schizophrenia, 105 in treatment of depression, increased bleeding risk
in treatment of secondary 74, 75, 85, 86 associated with, 79, 80,
mania, 95 in treatment of GAD, 142, 81
rivaroxaban, 80 143, 144 in pain management, 55
rivastigmine in treatment of panic potential side effects, 156
in treatment of Alzheimer’s disorder, 149 in treatment of anxiety,
disease, 34, 35 in treatment of PBA, 190 154–155
in treatment of agitation, serum electrolytes, 132 in treatment of depression,
26 serum levels, 86 75
in treatment of DLB, 18 measuring, 84–85, 86, in treatment of GAD,
romantic relationships, 90–91 142–143, 144
216–217 sexual expression, 117 in treatment of panic
teaching points, 217–219 in those with MNCD, 184, disorder, 149
ropinorole, 185 187 in treatment of PTSD, 164
sexuality, 202, 216–217 socialization, 144
schema-focused therapy, 180 take-home points, 204, spiritual needs, 46
schizoaffective disorder, 217–219 SSRIs
126–127 teaching points, 202–204, anxiety, and, 154
take-home points, 128 217–219 apathy, and, 30
teaching points, 127–128 sexually inappropriate delirium, and, 131
schizoid personality disorder, behavior (SIB), increased bleeding risk
176 183–184 associated with, 79–80
schizophrenia, 103–104, Alzheimer’s disease, and, potential side effects, 79,
See also treatment- 203 156
resistant schizophrenia definitions, 203 in treatment of anxiety,
take-home points, 104–105 depression, and, 86 154–155
teaching points, 104 evaluation and treatment, in treatment of depression,
schizotypal personality 186 74–75, 76, 79, 81, 85
disorder, 176 MNCDs, and, 204 in treatment of GAD,
sclopolamine, 131 nonpharmacologic 142–143, 144
SDRIs, 75 interventions, 185 in treatment of panic
seasonal affective disorder obtaining a history, 185 disorder, 147, 149
(SAD), 76 take-home points, 187 in treatment of PBA, 190
secondary mania, 94–95 teaching points, 184–186 in treatment of PTSD, 164
potential causes in older SIB, See sexually inappropriate in treatment of SIB, 186
adults, 95 behavior staff education
steps in treatment in long- situation-specific anxiety, on rights of long-term care
term care setting, 96 155 residents, 218
Index 231