Welcome to Current Psychiatry, a leading source of information, online and in print, for practitioners of psychiatry and its related subspecialties, including addiction psychiatry, child and adolescent psychiatry, and geriatric psychiatry. This Web site contains evidence-based reviews of the prevention, diagnosis, and treatment of mental illness and psychological disorders; case reports; updates on psychopharmacology; news about the specialty of psychiatry; pearls for practice; and other topics of interest and use to this audience.

Top Sections
Evidence-Based Reviews
Cases That Test Your Skills
cp

Dear Drupal User: You're seeing this because you're logged in to Drupal, and not redirected to MDedge.com/psychiatry. 

Main menu
CP Main Menu
Explore menu
CP Explore Menu
Proclivity ID
18803001
Unpublish
Negative Keywords
Bipolar depression
Depression
adolescent depression
adolescent major depressive disorder
adolescent schizophrenia
adolescent with major depressive disorder
animals
autism
baby
brexpiprazole
child
child bipolar
child depression
child schizophrenia
children with bipolar disorder
children with depression
children with major depressive disorder
compulsive behaviors
cure
elderly bipolar
elderly depression
elderly major depressive disorder
elderly schizophrenia
elderly with dementia
first break
first episode
gambling
gaming
geriatric depression
geriatric major depressive disorder
geriatric schizophrenia
infant
kid
major depressive disorder
major depressive disorder in adolescents
major depressive disorder in children
parenting
pediatric
pediatric bipolar
pediatric depression
pediatric major depressive disorder
pediatric schizophrenia
pregnancy
pregnant
rexulti
skin care
teen
wine
Negative Keywords Excluded Elements
header[@id='header']
section[contains(@class, 'nav-hidden')]
footer[@id='footer']
div[contains(@class, 'pane-pub-article-current-psychiatry')]
div[contains(@class, 'pane-pub-home-current-psychiatry')]
div[contains(@class, 'pane-pub-topic-current-psychiatry')]
div[contains(@class, 'panel-panel-inner')]
div[contains(@class, 'pane-node-field-article-topics')]
section[contains(@class, 'footer-nav-section-wrapper')]
Altmetric
DSM Affiliated
Display in offset block
QuickLearn Excluded Topics/Sections
Best Practices
CME
CME Supplements
Education Center
Medical Education Library
Disqus Exclude
Best Practices
CE/CME
Education Center
Medical Education Library
Enable Disqus
Display Author and Disclosure Link
Publication Type
Clinical
Slot System
Top 25
Disable Sticky Ads
Disable Ad Block Mitigation
Featured Buckets Admin
Publication LayerRX Default ID
798,799
Show Ads on this Publication's Homepage
Consolidated Pub
Show Article Page Numbers on TOC
Expire Announcement Bar
Mon, 04/29/2024 - 01:03
Use larger logo size
Off
publication_blueconic_enabled
Off
Show More Destinations Menu
Disable Adhesion on Publication
Off
Restore Menu Label on Mobile Navigation
Disable Facebook Pixel from Publication
Exclude this publication from publication selection on articles and quiz
Challenge Center
Disable Inline Native ads
survey writer start date
Mon, 04/29/2024 - 01:03
Current Issue
Title
Current Psychiatry
Description

Practical, evidence-based, peer-reviewed solutions to common clinical problems in psychiatry

Current Issue Reference

Breastfeeding by patients with serious mental illness: An ethical approach

Article Type
Changed
Tue, 12/12/2023 - 11:40
Display Headline
Breastfeeding by patients with serious mental illness: An ethical approach

Difficult ethical situations can arise when treating perinatal women who have serious mental illness (SMI). Clinicians must consider ethical issues related to administering antipsychotic medications, the safety of breastfeeding, and concerns for child welfare. They need to carefully weigh the risks and benefits of each decision when treating perinatal women who have SMI. Ethical guidelines can help clinicians best support families in these situations.

In this article, we describe 2 cases of women with psychotic disorders who requested to breastfeed after delivering their child during an inpatient psychiatric hospitalization. The course of their hospitalizations illustrated common ethical questions and facilitated the creation of a framework to assist with complex decision-making regarding breastfeeding on inpatient psychiatric units.

CASE 1

Ms. C, age 41, is multigravida with a psychiatric history of chronic, severe schizoaffective disorder and lives in supportive housing. When Ms. C presents to the hospital in search of a rape kit, clinicians discover she is 22 weeks pregnant but has not received any prenatal care. Psychiatry is consulted because she is found to be intermittently agitated and endorses grandiose delusions. Ms. C requires involuntary hospitalization for decompensated psychosis because she refuses prenatal and psychiatric care. Because it has reassuring reproductive safety data,1 olanzapine 5 mg/d is started. However, Ms. C experiences minimal improvement from a maximum dose of 20 mg/d. After 13 weeks on the psychiatry unit, she is transferred to obstetrics service for preeclampsia with severe features. Ms. C requires an urgent cesarean delivery at 37 weeks. Her baby boy is transferred to the neonatal intensive care unit (NICU) for transient tachypnea. After delivery and in consultation with psychiatry, the pediatrics team calls Child Protective Services (CPS) due to concern for neglect driven by Ms. C’s psychiatric condition. Ms. C visits the child with medical unit staff supervision in the NICU without consulting with the psychiatry service or CPS. On postpartum Day 2, Ms. C is transferred back to psychiatry for persistent psychosis.

On postpartum Day 3, Ms. C starts to produce breastmilk and requests to breastfeed. At this time, the multidisciplinary team determines she is not able to visit her child in the NICU due to psychiatric instability. No plan is developed to facilitate hand expression or pumping of breastmilk while Ms. C is on the psychiatric unit. The clinical teams discuss whether the benefits of breastfeeding and/or pumping breastmilk would outweigh the risks. CPS determines that Ms. C is unable to retain custody and places the child in kinship foster care while awaiting clinical improvement from her.

CASE 2

Ms. S, age 32, has a history of schizophrenia. She lives with her husband and parents. She is pregnant for the first time and has been receiving consistent prenatal care. Ms. S is brought to the hospital by her husband for bizarre behavior and paranoia after self-discontinuing risperidone 2 mg twice daily due to concern about the medication’s influence on her pregnancy. An ultrasound confirms she is 37 weeks pregnant. Psychiatry is consulted because Ms. S is internally preoccupied, delusional, and endorses auditory hallucinations. She requires involuntary hospitalization for decompensated psychosis. During admission, Ms. S experiences improvement of her psychiatric symptoms while receiving risperidone 2 mg twice daily, which she takes consistently after receiving extensive psychoeducation regarding its safety profile during pregnancy and lactation.

After 2 weeks on the psychiatry unit, Ms. S’s care team transfers her to the obstetrics service with one-to-one supervision. At 39 weeks gestation, she has a vaginal delivery without complications. Because there are no concerns about infant harm, obstetrics, pediatrics, and psychiatry coordinate care so the baby can room in with Ms. S, her husband, and a staff supervisor to facilitate bonding. Ms. S starts to lactate, wishes to breastfeed, and meets with lactation, pediatric, obstetric, and psychiatric specialists to discuss the risks and benefits of breastfeeding and pumping breastmilk. She pursues direct breastfeeding until the baby is discharged home with the husband at postpartum Day 2. CPS is not called because there are no concerns for parental abuse or neglect at the infant’s discharge.

On postpartum Day 2, the obstetrics service transfers Ms. S back to the psychiatric unit for further treatment of her paranoia. She wishes to pump breastmilk while hospitalized, so the treatment team supplies a breast pump, facilitates the storage of breastmilk, and coordinates supervision during pumping to reduce the ligature risk. Ms. S’s husband visits daily to transport the milk and feed the infant breastmilk and formula to meet its nutritional needs. Ms. S maintains psychiatric stability while breast pumping, and the team helps transition her to breastfeeding during visitation with her husband and infant until she is discharged home at 2 weeks postpartum.

Continue to: Approaching care with a relational ethics framework

 

 

Approaching care with a relational ethics framework

A relational ethics framework was constructed to evaluate whether to support breastfeeding for both patients during their psychiatric hospitalizations. A relational ethics perspective is defined as “a moral responsibility within a context of human relations” [that] “recognizes the human interdependency and reciprocity within which personal autonomy is embedded.”2 This framework values connectedness and commonality between various and even conflicting parties. In the setting of a clinician-patient relationship, health care decisions are made with consideration of the patient’s traditional beliefs, values, and principles rather than the application of impartial moral principles. For these complex cases, this framework was chosen to determine the safest possible outcome for both mother and child.

Risks/benefits of breastfeeding by patients who have SMI

There are several methods of breastfeeding, including direct breastfeeding and other ways of expressing breastmilk such as pumping or hand expression.3 Unlike other forms of feeding using breastmilk, direct breastfeeding has been extensively studied, has well-established medical and psychological benefits for newborns and mothers, and enhances long-term bonding.4 Compared with their counterparts who do not breastfeed, mothers who breastfeed have lower rates of unintended pregnancy, cardiovascular disease, postpartum bleeding, osteoporosis, and breast and ovarian cancer.5 Among its key psychological benefits, breastfeeding is associated with an increase in maternal self-efficacy and, in some research, has been shown to be associated with a decreased risk of postpartum depression and stress.Additionally, breastfed infants experience lower rates of childhood infection and obesity, and improved nutrition, cognitive development, and immune function.6 The American Academy of Pediatrics recognizes these benefits and recommends that women exclusively breastfeed for 6 months postpartum and continue to breastfeed for 2 years or beyond if mutually desired by the mother and child.7 Absolute contraindications to breastfeeding must be ruled out (eg, infant classic galactosemia; maternal use of illicit substances such as cocaine, opioids, or phencyclidine; maternal HIV infection, etc).

The risks of breastfeeding by patients who have SMI must also be considered. In severe situations, the infant can be exposed to a mother’s agitation secondary to psychosis.8,9 The transmission of antipsychotic medication through breastmilk and associated adverse effects (eg, sedation, poor feeding, and extrapyramidal symptoms) are also potential risks and varies among different antipsychotic medications.1,10 Therefore, when prescribing an antipsychotic for a patient with SMI who breastfeeds, it is crucial to consider the medication’s safety profile as well as other factors, such as the relative infant dose (the weight-adjusted [ie, mg/kg] percentage of the maternal dosage ingested by a fully breastfed infant) and the molecular characteristics of the medication.10-12 Neonates should be routinely monitored for adverse effects, medication toxicity, and withdrawal symptoms, and care should be coordinated with the infant’s pediatrician. Certain antipsychotic medications, such as aripiprazole, may impact breastmilk production through the dopamine agonist’s interference of the prolactin reflex and anticholinergic properties.11,13 For a patient with SMI, perhaps the most significant risk involves the time and resources needed for breastfeeding, which can interfere with sleep and psychiatric treatment and possibly further exacerbate psychiatric symptoms.14-16 Additionally, breastfeeding difficulties or disruption can increase the risk of psychiatric symptoms and psychological distress.17 In Ms. C’s case, there was a delay in the baby latching as well as multiple medical and psychiatric factors that hindered the milk-ejection reflex to properly initiate; both of these factors rendered breastfeeding particularly difficult while Ms. C was on the inpatient psychiatry unit.17 In comparison, Ms. S was able to bond with her infant shortly after delivery, which facilitated the milk-ejection reflex and lactation.

Patients who wish to directly breastfeed but struggle to do so while tending to their acute psychiatric condition can benefit from expression of breastmilk that can be provided to the infant or discarded to facilitate breastfeeding in the future.18 While expression of breastmilk may not be as advantageous for infant health as direct breastfeeding due to the potential changes in breastmilk composition from collecting, storing, and heating, this option can be more protective than formula feeding and facilitate future breastfeeding.19 In these clinical scenarios, it is standard care to provide a hospital-grade breast pump to the patient, much like a continuous positive airway pressure machine is provided to patients with obstructive sleep apnea.20 However, there is often considerable difficulty obtaining proper breastfeeding equipment and a lack of services devoted to perinatal care in general inpatient settings. Barriers to direct breastfeeding and pumping of breastmilk are highlighted in the Table.21

CP02212e16_t1.png

Limitations on breastfeeding on an inpatient unit

The limitations in care and restrictions placed on breastfeeding are more optimally addressed in a mother and baby unit (MBU). MBUs are specialized inpatient psychiatric units designed for mothers experiencing severe perinatal psychiatric difficulties. Unlike general psychiatric units, MBUs allow for joint, full-time admission of mothers and their infants. These units also include multidisciplinary staff who specialize in treating perinatal mental health issues as well as infant care and child development.22 Admission into an MBU is considered best practice for new mothers requiring treatment, particularly in the United Kingdom, Australia, and France, as it is well-recognized that the separation of mother and baby can be psychologically harmful.23 In the UK, most patients admitted to an MBU showed significant improvement of their psychiatric symptoms and reported overall high satisfaction with care.24,25 Patients who experience postpartum psychosis prefer MBUs over general psychiatric units because the latter often lack specialized perinatal support, appropriate visitor arrangements, and adequate time with their infant.26-28

Continue to: The resistance to adopting MBUs in the United States...

 

 

The resistance to adopting MBUs in the United States has posed significant barriers in care for perinatal patients and has been attributed to financial barriers, medicolegal risk, staffing, and safety concerns.29 Though currently there are no MBUs in the US, other specialized units have been created. A partial day hospitalization program created in 2000 in Rhode Island for mothers and infants revolutionized the psychiatric care experience for new mothers.30 Since then, other institutions have significantly expanded their services to include perinatal psychiatry inpatient units, yet unlike MBUs, these units typically do not provide overnight rooming-in with infants.31 They have the necessary resources and facilities to accommodate the mother’s needs and maximize positive mother-infant interaction, while actively integrating the infant into the mother’s treatment. Breast pumping is treated as a necessary medical procedure and patients can easily access hospital-grade breast pumps with staff supervision. At one such perinatal psychiatric inpatient unit, high rates of treatment satisfaction and significant improvements in symptoms of depression, anxiety, active suicidal ideation, and overall functioning were observed at discharge.32 Therefore, it is crucial to incorporate strategies in general psychiatry units to improve perinatal care, acknowledging that most patients will not have access to these specialized units.21

A framework to approaching the relational ethics decisions

An interdisciplinary team used a relational ethics perspective to carefully analyze the risks and benefits of these complex cases. In Figure 1, we propose a framework for the relational ethics decisions of breastfeeding on general inpatient psychiatric units. In creating this framework, we considered principles of autonomy, beneficence, and nonmaleficence, along with the medical and logistical barriers to breastfeeding.

CP02212e16_f1.png

In Ms. C’s case, the team determined that the risks—which included disrupting the mother’s psychiatric treatment, exposing her to psychological harm due to increasing attachment before remanding the child to CPS custody, and risks to the child due to potential unpredictable agitation driven by the treatment-refractory psychosis of the mother as well as that of other psychiatric patients—outweighed the benefits of breastfeeding. We instead recommended breast pumping as an alternative once Ms. C’s psychiatric stability improved. We presented Ms. C with the option of breast pumping on postpartum Day 5. During a 1-day period in which she showed improved behavioral control, she was counseled on the risks and benefits of breastfeeding and exclusive pumping and was notified that the team would help her with the necessary resources, including consultation with a lactation specialist and breast pump. Despite lactation consultant support, Ms. C had low milk production and difficulty with hand expression, which was very discouraging to her. She produced 1 ounce of milk that was shared with the newborn while in the NICU. Because Ms. C’s psychiatric symptoms continued to be severe, with lability and aggression, and because pumping was triggering distress, the multidisciplinary team determined the best course of care would be to focus on her psychiatric recovery rather than on pumping breastmilk. To reduce milk production and minimize discomfort secondary to breast engorgement, the lactation consultant recommended cold compresses, pain management, and compression of breasts. Ultimately, the mother-infant dyad was unable to reap the benefits of breastfeeding (via pumping or direct breastfeeding) due to the mother’s underlying psychiatric illness, although the staffing, psychosocial support, and logistical limitations contributed to this outcome.33

In Ms. S’s case, the treatment team determined that there were no medical or psychiatric contraindications to breastfeeding, and she was counseled on the risks and benefits of direct breastfeeding and pumping. The treatment team determined it was safe for Ms. S to directly breastfeed as there were no concerns for infant harm post­delivery with constant supervision while on the obstetrics floor. The patient opted to directly breastfeed, which was successful with the guidance of a lactation specialist. When she was transferred to the psychiatric unit on postpartum Day 2, her child was discharged home with the husband. The patient was then encouraged to pump while the psychiatrists monitored her symptoms closely and facilitated increased staff and resources. Transportation of breastmilk was made possible by the family, and on postpartum Day 5, as the patient maintained psychiatric stability, the team discussed with Ms. S and her husband the prospect of direct breastfeeding. The treatment team arranged for separate visitation hours to minimize the possibility of exposing the infant to aggression from other patients on the unit and advocated with hospital leadership to approve of infant visitation on the unit.

Impact of involvement of Child Protective Services

The involvement of CPS also added complexity to Ms. C’s case. Without proper legal guidance, mothers with psychosis who lose custody can find it difficult to navigate the legal system and maintain contact with their children.34 As the prevalence of custody loss in mothers with psychosis is high (approximately 50% according to research published in the last 10 years), effective interventions to reunite the mother and child must be promoted (Figure 2).35-39 Ultimately, the goal of psychiatric hospitalization for perinatal women who have SMI is psychiatric stabilization. The preemptive involvement of psychiatry is crucial because it can allow for early postpartum planning and can provide an opportunity to address feeding options and custody concerns with the patient, social supports and services, and various medical teams. In Ms. C’s case, she visited her baby in the NICU on postpartum Day 2 without consultation with psychiatry or CPS, which posed risks to the patient, infant, and staff. It is vital that various clinicians collaborate with each other and the patient, working towards the goal of optimizing the patient’s mental health to allow for parenting rights in the future and maximizing a sustainable attachment between the parent and child. In Ms. S’s case, the husband was able to facilitate caring for the baby while the mother was hospitalized and played an integral role in the feeding process via pumped breastmilk and transport of the infant for direct breastfeeding.

CP02212e16_f2.png

Continue to: The differences in these 2 cases...

 

 

The differences in these 2 cases show the extreme importance of social support to benefit both the mother and child, and the need for more comprehensive social services for women who do not have a social safety net.

Bottom Line

These complex cases highlight an ethical decision-making approach to breastfeeding in perinatal women who have serious mental illness. Collaborative care and shared decision-making, which highlight the interests of the mother and baby, are crucial when assessing the risks and benefits of breastfeeding and pumping breastmilk. Our relational ethics framework can be used to better evaluate and implement breastfeeding options on general psychiatric units.

Related Resources

Drug Brand Names

Aripiprazole • Abilify
Olanzapine • Zyprexa
Risperidone • Risperdal

[embed:render:related:node:240620]

[embed:render:related:node:262580]

References

1. Brunner E, Falk DM, Jones M, et al. Olanzapine in pregnancy and breastfeeding: a review of data from global safety surveillance. BMC Pharmacol Toxicol. 2013;14:38. doi:10.1186/2050-6511-14-38

2. Seeman MV. Relational ethics: when mothers suffer from psychosis. Arch Womens Ment Health. 2004;7(3):201-210. doi:10.1007/s00737-004-0054-8

3. Motee A, Jeewon R. Importance of exclusive breastfeeding and complementary feeding among infants. Curr Res Nutr Food Sci. 2014;2(2). doi:10.12944/CRNFSJ.2.2.02

4. Committee Opinion No. 570: breastfeeding in underserved women: increasing initiation and continuation of breastfeeding. Obstet Gynecol. 2013;122(2 Pt 1):423-427. doi:10.1097/01.AOG.0000433008.93971.6a

5. Sibolboro Mezzacappa E, Endicott J. Parity mediates the association between infant feeding method and maternal depressive symptoms in the postpartum. Arch Womens Ment Health. 2007;10(6):259-266. doi:10.1007/s00737-007-0207-7

6. Kramer MS, Chalmers B, Hodnett ED, et al. Promotion of Breastfeeding Intervention Trial (PROBIT): a randomized trial in the Republic of Belarus. JAMA. 2001;285(4):413-420. doi:10.1001/jama.285.4.413

7. American Academy of Pediatrics. American Academy of Pediatrics calls for more support for breastfeeding mothers within updated policy recommendations. June 27, 2022. Accessed October 4, 2022. https://www.aap.org/en/news-room/news-releases/aap/2022/american-academy-of-pediatrics-calls-for-more-support-for-breastfeeding-mothers-within-updated-policy-recommendations

8. Hipwell AE, Kumar R. Maternal psychopathology and prediction of outcome based on mother-infant interaction ratings (BMIS). Br J Psychiatry. 1996;169(5):655-661. doi:10.1192/bjp.169.5.655

9. Chandra PS, Bhargavaraman RP, Raghunandan VN, et al. Delusions related to infant and their association with mother-infant interactions in postpartum psychotic disorders. Arch Womens Ment Health. 2006;9(5):285-288. doi:10.1007/s00737-006-0147-7

10. Klinger G, Stahl B, Fusar-Poli P, et al. Antipsychotic drugs and breastfeeding. Pediatr Endocrinol Rev. 2013;10(3):308-317.

11. Uguz F. A new safety scoring system for the use of psychotropic drugs during lactation. Am J Ther. 2021;28(1):e118-e126. doi:10.1097/MJT.0000000000000909

12. Hale TW, Krutsch K. Hale’s Medications & Mothers’ Milk, 2023: A Manual of Lactational Pharmacology. 20th ed. Springer Publishing Company; 2023.

13. Komaroff A. Aripiprazole and lactation failure: the importance of shared decision making. A case report. Case Rep Womens Health. 2021;30:e00308. doi:10.1016/j.crwh.2021.e00308‌

14. Dennis CL, McQueen K. Does maternal postpartum depressive symptomatology influence infant feeding outcomes? Acta Pediatr. 2007;96(4):590-594. doi:10.1111/j.1651-2227.2007.00184.x

15. Chaput KH, Nettel-Aguirre A, Musto R, et al. Breastfeeding difficulties and supports and risk of postpartum depression in a cohort of women who have given birth in Calgary: a prospective cohort study. CMAJ Open. 2016;4(1):E103-E109. doi:10.9778/cmajo.20150009

16. Dias CC, Figueiredo B. Breastfeeding and depression: a systematic review of the literature. J Affect Disord. 2015;171:142-154. doi:10.1016/j.jad.2014.09.022

17. Brown A, Rance J, Bennett P. Understanding the relationship between breastfeeding and postnatal depression: the role of pain and physical difficulties. J Adv Nurs. 2016;72(2):273-282. doi:10.1111/jan.12832

18. Rosenbaum KA. Exclusive breastmilk pumping: a concept analysis. Nurs Forum. 2022;57(5):946-953. doi:10.1111/nuf.12766

19. Boone KM, Geraghty SR, Keim SA. Feeding at the breast and expressed milk feeding: associations with otitis media and diarrhea in infants. J Pediatr. 2016;174:118-125. doi:10.1016/j.jpeds.2016.04.006

20. Epstein LJ, Kristo D, Strollo PJ Jr, et al; Adult Obstructive Sleep Apnea Task Force of the American Academy of Sleep Medicine. Clinical guideline for the evaluation, management and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med. 2009;5(3):263-276.

21. Caan MP, Sreshta NE, Okwerekwu JA, et al. Clinical and legal considerations regarding breastfeeding on psychiatric units. J Am Acad Psychiatry Law. 2022;50(2):200-207. doi:10.29158/JAAPL.210086-21

22. Glangeaud-Freudenthal NMC, Rainelli C, Cazas O, et al. Inpatient mother and baby psychiatric units (MBUs) and day cares. In: Sutter-Dallay AL, Glangeaud-Freudenthal NC, Guedeney A, et al, eds. Joint Care of Parents and Infants in Perinatal Psychiatry. Springer, Cham; 2016:147-164. doi:10.1007/978-3-319-21557-0_10

23. Dembosky A. A humane approach to caring for new mothers in psychiatric crisis. Health Aff (Millwood). 2021;40(10):1528-1533. doi:10.1377/hlthaff.2021.01288

24. Connellan K, Bartholomaeus C, Due C, et al. A systematic review of research on psychiatric mother-baby units. Arch Womens Ment Health. 2017;20(3):373-388. doi:10.1007/s00737-017-0718-9

25. Griffiths J, Lever Taylor B, Morant N, et al. A qualitative comparison of experiences of specialist mother and baby units versus general psychiatric wards. BMC Psychiatry. 2019;19(1):401. doi:10.1186/s12888-019-2389-8

26. Heron J, Gilbert N, Dolman C, et al. Information and support needs during recovery from postpartum psychosis. Arch Womens Ment Health. 2012;15(3):155-165. doi:10.1007/s00737-012-0267-1

27. Robertson E, Lyons A. Living with puerperal psychosis: a qualitative analysis. Psychol Psychother. 2003;76(Pt 4):411-431. doi:10.1348/147608303770584755

28. Mental Welfare Commission for Scotland. Perinatal Themed Visit Report: Keeping Mothers and Babies in Mind. Mental Welfare Commission for Scotland; 2016.

29. Wisner KL, Jennings KD, Conley B. Clinical dilemmas due to the lack of inpatient mother-baby units. Int J Psychiatry Med. 1996;26(4):479-493. doi:10.2190/NFJK-A4V7-CXUU-AM89

30. Battle CL, Howard MM. A mother-baby psychiatric day hospital: history, rationale, and why perinatal mental health is important for obstetric medicine. Obstet Med. 2014;7(2):66-70. doi:10.1177/1753495X13514402

31. Bullard ES, Meltzer-Brody S, Rubinow DR. The need for comprehensive psychiatric perinatal care-the University of North Carolina at Chapel Hill, Department of Psychiatry, Center for Women’s Mood Disorders launches the first dedicated inpatient program in the United States. Am J Obstet Gynecol. 2009;201(5):e10-e11. doi:10.1016/j.ajog.2009.05.004

32. Meltzer-Brody S, Brandon AR, Pearson B, et al. Evaluating the clinical effectiveness of a specialized perinatal psychiatry inpatient unit. Arch Womens Ment Health. 2014;17(2):107-113. doi:10.1007/s00737-013-0390-7

33. Alvarez-Toro V. Gender-specific care for women in psychiatric units. J Am Acad Psychiatry Law. 2022;JAAPL.220015-21. doi:10.29158/JAAPL.220015-21

34. Diaz-Caneja A, Johnson S. The views and experiences of severely mentally ill mothers--a qualitative study. Soc Psychiatry Psychiatr Epidemiol. 2004;39(6):472-482. doi:10.1007/s00127-004-0772-2

35. Gewurtz R, Krupa T, Eastabrook S, et al. Prevalence and characteristics of parenting among people served by assertive community treatment. Psychiatr Rehabil J. 2004;28(1):63-65. doi:10.2975/28.2004.63.65

36. Howard LM, Kumar R, Thornicroft G. Psychosocial characteristics and needs of mothers with psychotic disorders. Br J Psychiatry. 2001;178:427-432. doi:10.1192/bjp.178.5.427

37. Hollingsworth LD. Child custody loss among women with persistent severe mental illness. Social Work Research. 2004;28(4):199-209. doi:10.1093/swr/28.4.199

38. Dipple H, Smith S, Andrews H, et al. The experience of motherhood in women with severe and enduring mental illness. Soc Psychiatry Psychiatr Epidemiolf. 2002;37(7):336-340. doi:10.1007/s00127-002-0559-2

39. Seeman MV. Intervention to prevent child custody loss in mothers with schizophrenia. Schizophr Res Treatment. 2012;2012:796763. doi:10.1155/2012/796763

Article PDF
Author and Disclosure Information

Sitara Soundararajan, MD
PGY-3 Psychiatry Resident
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center
Bronx, New York

Marlee Madora, MD
Women’s Mental Health Fellow
Department of Psychiatry
Brigham and Women’s Hospital
Boston, Massachusetts

Rubiahna Vaughn, MD, MPH
Assistant Professor
Department of Psychiatry and Behavioral Services
Albert Einstein College of Medicine
Director, Consultation-Liaison and Emergency Psychiatry
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center, Weiler Hospital
Bronx, New York

Sarah Kauffman, MD
Attending Psychiatrist
The Motherhood Center of New York
New York, New York

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Issue
Current Psychiatry - 22(12)
Publications
Topics
Page Number
e16-e23
Sections
Author and Disclosure Information

Sitara Soundararajan, MD
PGY-3 Psychiatry Resident
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center
Bronx, New York

Marlee Madora, MD
Women’s Mental Health Fellow
Department of Psychiatry
Brigham and Women’s Hospital
Boston, Massachusetts

Rubiahna Vaughn, MD, MPH
Assistant Professor
Department of Psychiatry and Behavioral Services
Albert Einstein College of Medicine
Director, Consultation-Liaison and Emergency Psychiatry
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center, Weiler Hospital
Bronx, New York

Sarah Kauffman, MD
Attending Psychiatrist
The Motherhood Center of New York
New York, New York

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Author and Disclosure Information

Sitara Soundararajan, MD
PGY-3 Psychiatry Resident
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center
Bronx, New York

Marlee Madora, MD
Women’s Mental Health Fellow
Department of Psychiatry
Brigham and Women’s Hospital
Boston, Massachusetts

Rubiahna Vaughn, MD, MPH
Assistant Professor
Department of Psychiatry and Behavioral Services
Albert Einstein College of Medicine
Director, Consultation-Liaison and Emergency Psychiatry
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center, Weiler Hospital
Bronx, New York

Sarah Kauffman, MD
Attending Psychiatrist
The Motherhood Center of New York
New York, New York

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Article PDF
Article PDF

Difficult ethical situations can arise when treating perinatal women who have serious mental illness (SMI). Clinicians must consider ethical issues related to administering antipsychotic medications, the safety of breastfeeding, and concerns for child welfare. They need to carefully weigh the risks and benefits of each decision when treating perinatal women who have SMI. Ethical guidelines can help clinicians best support families in these situations.

In this article, we describe 2 cases of women with psychotic disorders who requested to breastfeed after delivering their child during an inpatient psychiatric hospitalization. The course of their hospitalizations illustrated common ethical questions and facilitated the creation of a framework to assist with complex decision-making regarding breastfeeding on inpatient psychiatric units.

CASE 1

Ms. C, age 41, is multigravida with a psychiatric history of chronic, severe schizoaffective disorder and lives in supportive housing. When Ms. C presents to the hospital in search of a rape kit, clinicians discover she is 22 weeks pregnant but has not received any prenatal care. Psychiatry is consulted because she is found to be intermittently agitated and endorses grandiose delusions. Ms. C requires involuntary hospitalization for decompensated psychosis because she refuses prenatal and psychiatric care. Because it has reassuring reproductive safety data,1 olanzapine 5 mg/d is started. However, Ms. C experiences minimal improvement from a maximum dose of 20 mg/d. After 13 weeks on the psychiatry unit, she is transferred to obstetrics service for preeclampsia with severe features. Ms. C requires an urgent cesarean delivery at 37 weeks. Her baby boy is transferred to the neonatal intensive care unit (NICU) for transient tachypnea. After delivery and in consultation with psychiatry, the pediatrics team calls Child Protective Services (CPS) due to concern for neglect driven by Ms. C’s psychiatric condition. Ms. C visits the child with medical unit staff supervision in the NICU without consulting with the psychiatry service or CPS. On postpartum Day 2, Ms. C is transferred back to psychiatry for persistent psychosis.

On postpartum Day 3, Ms. C starts to produce breastmilk and requests to breastfeed. At this time, the multidisciplinary team determines she is not able to visit her child in the NICU due to psychiatric instability. No plan is developed to facilitate hand expression or pumping of breastmilk while Ms. C is on the psychiatric unit. The clinical teams discuss whether the benefits of breastfeeding and/or pumping breastmilk would outweigh the risks. CPS determines that Ms. C is unable to retain custody and places the child in kinship foster care while awaiting clinical improvement from her.

CASE 2

Ms. S, age 32, has a history of schizophrenia. She lives with her husband and parents. She is pregnant for the first time and has been receiving consistent prenatal care. Ms. S is brought to the hospital by her husband for bizarre behavior and paranoia after self-discontinuing risperidone 2 mg twice daily due to concern about the medication’s influence on her pregnancy. An ultrasound confirms she is 37 weeks pregnant. Psychiatry is consulted because Ms. S is internally preoccupied, delusional, and endorses auditory hallucinations. She requires involuntary hospitalization for decompensated psychosis. During admission, Ms. S experiences improvement of her psychiatric symptoms while receiving risperidone 2 mg twice daily, which she takes consistently after receiving extensive psychoeducation regarding its safety profile during pregnancy and lactation.

After 2 weeks on the psychiatry unit, Ms. S’s care team transfers her to the obstetrics service with one-to-one supervision. At 39 weeks gestation, she has a vaginal delivery without complications. Because there are no concerns about infant harm, obstetrics, pediatrics, and psychiatry coordinate care so the baby can room in with Ms. S, her husband, and a staff supervisor to facilitate bonding. Ms. S starts to lactate, wishes to breastfeed, and meets with lactation, pediatric, obstetric, and psychiatric specialists to discuss the risks and benefits of breastfeeding and pumping breastmilk. She pursues direct breastfeeding until the baby is discharged home with the husband at postpartum Day 2. CPS is not called because there are no concerns for parental abuse or neglect at the infant’s discharge.

On postpartum Day 2, the obstetrics service transfers Ms. S back to the psychiatric unit for further treatment of her paranoia. She wishes to pump breastmilk while hospitalized, so the treatment team supplies a breast pump, facilitates the storage of breastmilk, and coordinates supervision during pumping to reduce the ligature risk. Ms. S’s husband visits daily to transport the milk and feed the infant breastmilk and formula to meet its nutritional needs. Ms. S maintains psychiatric stability while breast pumping, and the team helps transition her to breastfeeding during visitation with her husband and infant until she is discharged home at 2 weeks postpartum.

Continue to: Approaching care with a relational ethics framework

 

 

Approaching care with a relational ethics framework

A relational ethics framework was constructed to evaluate whether to support breastfeeding for both patients during their psychiatric hospitalizations. A relational ethics perspective is defined as “a moral responsibility within a context of human relations” [that] “recognizes the human interdependency and reciprocity within which personal autonomy is embedded.”2 This framework values connectedness and commonality between various and even conflicting parties. In the setting of a clinician-patient relationship, health care decisions are made with consideration of the patient’s traditional beliefs, values, and principles rather than the application of impartial moral principles. For these complex cases, this framework was chosen to determine the safest possible outcome for both mother and child.

Risks/benefits of breastfeeding by patients who have SMI

There are several methods of breastfeeding, including direct breastfeeding and other ways of expressing breastmilk such as pumping or hand expression.3 Unlike other forms of feeding using breastmilk, direct breastfeeding has been extensively studied, has well-established medical and psychological benefits for newborns and mothers, and enhances long-term bonding.4 Compared with their counterparts who do not breastfeed, mothers who breastfeed have lower rates of unintended pregnancy, cardiovascular disease, postpartum bleeding, osteoporosis, and breast and ovarian cancer.5 Among its key psychological benefits, breastfeeding is associated with an increase in maternal self-efficacy and, in some research, has been shown to be associated with a decreased risk of postpartum depression and stress.Additionally, breastfed infants experience lower rates of childhood infection and obesity, and improved nutrition, cognitive development, and immune function.6 The American Academy of Pediatrics recognizes these benefits and recommends that women exclusively breastfeed for 6 months postpartum and continue to breastfeed for 2 years or beyond if mutually desired by the mother and child.7 Absolute contraindications to breastfeeding must be ruled out (eg, infant classic galactosemia; maternal use of illicit substances such as cocaine, opioids, or phencyclidine; maternal HIV infection, etc).

The risks of breastfeeding by patients who have SMI must also be considered. In severe situations, the infant can be exposed to a mother’s agitation secondary to psychosis.8,9 The transmission of antipsychotic medication through breastmilk and associated adverse effects (eg, sedation, poor feeding, and extrapyramidal symptoms) are also potential risks and varies among different antipsychotic medications.1,10 Therefore, when prescribing an antipsychotic for a patient with SMI who breastfeeds, it is crucial to consider the medication’s safety profile as well as other factors, such as the relative infant dose (the weight-adjusted [ie, mg/kg] percentage of the maternal dosage ingested by a fully breastfed infant) and the molecular characteristics of the medication.10-12 Neonates should be routinely monitored for adverse effects, medication toxicity, and withdrawal symptoms, and care should be coordinated with the infant’s pediatrician. Certain antipsychotic medications, such as aripiprazole, may impact breastmilk production through the dopamine agonist’s interference of the prolactin reflex and anticholinergic properties.11,13 For a patient with SMI, perhaps the most significant risk involves the time and resources needed for breastfeeding, which can interfere with sleep and psychiatric treatment and possibly further exacerbate psychiatric symptoms.14-16 Additionally, breastfeeding difficulties or disruption can increase the risk of psychiatric symptoms and psychological distress.17 In Ms. C’s case, there was a delay in the baby latching as well as multiple medical and psychiatric factors that hindered the milk-ejection reflex to properly initiate; both of these factors rendered breastfeeding particularly difficult while Ms. C was on the inpatient psychiatry unit.17 In comparison, Ms. S was able to bond with her infant shortly after delivery, which facilitated the milk-ejection reflex and lactation.

Patients who wish to directly breastfeed but struggle to do so while tending to their acute psychiatric condition can benefit from expression of breastmilk that can be provided to the infant or discarded to facilitate breastfeeding in the future.18 While expression of breastmilk may not be as advantageous for infant health as direct breastfeeding due to the potential changes in breastmilk composition from collecting, storing, and heating, this option can be more protective than formula feeding and facilitate future breastfeeding.19 In these clinical scenarios, it is standard care to provide a hospital-grade breast pump to the patient, much like a continuous positive airway pressure machine is provided to patients with obstructive sleep apnea.20 However, there is often considerable difficulty obtaining proper breastfeeding equipment and a lack of services devoted to perinatal care in general inpatient settings. Barriers to direct breastfeeding and pumping of breastmilk are highlighted in the Table.21

CP02212e16_t1.png

Limitations on breastfeeding on an inpatient unit

The limitations in care and restrictions placed on breastfeeding are more optimally addressed in a mother and baby unit (MBU). MBUs are specialized inpatient psychiatric units designed for mothers experiencing severe perinatal psychiatric difficulties. Unlike general psychiatric units, MBUs allow for joint, full-time admission of mothers and their infants. These units also include multidisciplinary staff who specialize in treating perinatal mental health issues as well as infant care and child development.22 Admission into an MBU is considered best practice for new mothers requiring treatment, particularly in the United Kingdom, Australia, and France, as it is well-recognized that the separation of mother and baby can be psychologically harmful.23 In the UK, most patients admitted to an MBU showed significant improvement of their psychiatric symptoms and reported overall high satisfaction with care.24,25 Patients who experience postpartum psychosis prefer MBUs over general psychiatric units because the latter often lack specialized perinatal support, appropriate visitor arrangements, and adequate time with their infant.26-28

Continue to: The resistance to adopting MBUs in the United States...

 

 

The resistance to adopting MBUs in the United States has posed significant barriers in care for perinatal patients and has been attributed to financial barriers, medicolegal risk, staffing, and safety concerns.29 Though currently there are no MBUs in the US, other specialized units have been created. A partial day hospitalization program created in 2000 in Rhode Island for mothers and infants revolutionized the psychiatric care experience for new mothers.30 Since then, other institutions have significantly expanded their services to include perinatal psychiatry inpatient units, yet unlike MBUs, these units typically do not provide overnight rooming-in with infants.31 They have the necessary resources and facilities to accommodate the mother’s needs and maximize positive mother-infant interaction, while actively integrating the infant into the mother’s treatment. Breast pumping is treated as a necessary medical procedure and patients can easily access hospital-grade breast pumps with staff supervision. At one such perinatal psychiatric inpatient unit, high rates of treatment satisfaction and significant improvements in symptoms of depression, anxiety, active suicidal ideation, and overall functioning were observed at discharge.32 Therefore, it is crucial to incorporate strategies in general psychiatry units to improve perinatal care, acknowledging that most patients will not have access to these specialized units.21

A framework to approaching the relational ethics decisions

An interdisciplinary team used a relational ethics perspective to carefully analyze the risks and benefits of these complex cases. In Figure 1, we propose a framework for the relational ethics decisions of breastfeeding on general inpatient psychiatric units. In creating this framework, we considered principles of autonomy, beneficence, and nonmaleficence, along with the medical and logistical barriers to breastfeeding.

CP02212e16_f1.png

In Ms. C’s case, the team determined that the risks—which included disrupting the mother’s psychiatric treatment, exposing her to psychological harm due to increasing attachment before remanding the child to CPS custody, and risks to the child due to potential unpredictable agitation driven by the treatment-refractory psychosis of the mother as well as that of other psychiatric patients—outweighed the benefits of breastfeeding. We instead recommended breast pumping as an alternative once Ms. C’s psychiatric stability improved. We presented Ms. C with the option of breast pumping on postpartum Day 5. During a 1-day period in which she showed improved behavioral control, she was counseled on the risks and benefits of breastfeeding and exclusive pumping and was notified that the team would help her with the necessary resources, including consultation with a lactation specialist and breast pump. Despite lactation consultant support, Ms. C had low milk production and difficulty with hand expression, which was very discouraging to her. She produced 1 ounce of milk that was shared with the newborn while in the NICU. Because Ms. C’s psychiatric symptoms continued to be severe, with lability and aggression, and because pumping was triggering distress, the multidisciplinary team determined the best course of care would be to focus on her psychiatric recovery rather than on pumping breastmilk. To reduce milk production and minimize discomfort secondary to breast engorgement, the lactation consultant recommended cold compresses, pain management, and compression of breasts. Ultimately, the mother-infant dyad was unable to reap the benefits of breastfeeding (via pumping or direct breastfeeding) due to the mother’s underlying psychiatric illness, although the staffing, psychosocial support, and logistical limitations contributed to this outcome.33

In Ms. S’s case, the treatment team determined that there were no medical or psychiatric contraindications to breastfeeding, and she was counseled on the risks and benefits of direct breastfeeding and pumping. The treatment team determined it was safe for Ms. S to directly breastfeed as there were no concerns for infant harm post­delivery with constant supervision while on the obstetrics floor. The patient opted to directly breastfeed, which was successful with the guidance of a lactation specialist. When she was transferred to the psychiatric unit on postpartum Day 2, her child was discharged home with the husband. The patient was then encouraged to pump while the psychiatrists monitored her symptoms closely and facilitated increased staff and resources. Transportation of breastmilk was made possible by the family, and on postpartum Day 5, as the patient maintained psychiatric stability, the team discussed with Ms. S and her husband the prospect of direct breastfeeding. The treatment team arranged for separate visitation hours to minimize the possibility of exposing the infant to aggression from other patients on the unit and advocated with hospital leadership to approve of infant visitation on the unit.

Impact of involvement of Child Protective Services

The involvement of CPS also added complexity to Ms. C’s case. Without proper legal guidance, mothers with psychosis who lose custody can find it difficult to navigate the legal system and maintain contact with their children.34 As the prevalence of custody loss in mothers with psychosis is high (approximately 50% according to research published in the last 10 years), effective interventions to reunite the mother and child must be promoted (Figure 2).35-39 Ultimately, the goal of psychiatric hospitalization for perinatal women who have SMI is psychiatric stabilization. The preemptive involvement of psychiatry is crucial because it can allow for early postpartum planning and can provide an opportunity to address feeding options and custody concerns with the patient, social supports and services, and various medical teams. In Ms. C’s case, she visited her baby in the NICU on postpartum Day 2 without consultation with psychiatry or CPS, which posed risks to the patient, infant, and staff. It is vital that various clinicians collaborate with each other and the patient, working towards the goal of optimizing the patient’s mental health to allow for parenting rights in the future and maximizing a sustainable attachment between the parent and child. In Ms. S’s case, the husband was able to facilitate caring for the baby while the mother was hospitalized and played an integral role in the feeding process via pumped breastmilk and transport of the infant for direct breastfeeding.

CP02212e16_f2.png

Continue to: The differences in these 2 cases...

 

 

The differences in these 2 cases show the extreme importance of social support to benefit both the mother and child, and the need for more comprehensive social services for women who do not have a social safety net.

Bottom Line

These complex cases highlight an ethical decision-making approach to breastfeeding in perinatal women who have serious mental illness. Collaborative care and shared decision-making, which highlight the interests of the mother and baby, are crucial when assessing the risks and benefits of breastfeeding and pumping breastmilk. Our relational ethics framework can be used to better evaluate and implement breastfeeding options on general psychiatric units.

Related Resources

Drug Brand Names

Aripiprazole • Abilify
Olanzapine • Zyprexa
Risperidone • Risperdal

[embed:render:related:node:240620]

[embed:render:related:node:262580]

Difficult ethical situations can arise when treating perinatal women who have serious mental illness (SMI). Clinicians must consider ethical issues related to administering antipsychotic medications, the safety of breastfeeding, and concerns for child welfare. They need to carefully weigh the risks and benefits of each decision when treating perinatal women who have SMI. Ethical guidelines can help clinicians best support families in these situations.

In this article, we describe 2 cases of women with psychotic disorders who requested to breastfeed after delivering their child during an inpatient psychiatric hospitalization. The course of their hospitalizations illustrated common ethical questions and facilitated the creation of a framework to assist with complex decision-making regarding breastfeeding on inpatient psychiatric units.

CASE 1

Ms. C, age 41, is multigravida with a psychiatric history of chronic, severe schizoaffective disorder and lives in supportive housing. When Ms. C presents to the hospital in search of a rape kit, clinicians discover she is 22 weeks pregnant but has not received any prenatal care. Psychiatry is consulted because she is found to be intermittently agitated and endorses grandiose delusions. Ms. C requires involuntary hospitalization for decompensated psychosis because she refuses prenatal and psychiatric care. Because it has reassuring reproductive safety data,1 olanzapine 5 mg/d is started. However, Ms. C experiences minimal improvement from a maximum dose of 20 mg/d. After 13 weeks on the psychiatry unit, she is transferred to obstetrics service for preeclampsia with severe features. Ms. C requires an urgent cesarean delivery at 37 weeks. Her baby boy is transferred to the neonatal intensive care unit (NICU) for transient tachypnea. After delivery and in consultation with psychiatry, the pediatrics team calls Child Protective Services (CPS) due to concern for neglect driven by Ms. C’s psychiatric condition. Ms. C visits the child with medical unit staff supervision in the NICU without consulting with the psychiatry service or CPS. On postpartum Day 2, Ms. C is transferred back to psychiatry for persistent psychosis.

On postpartum Day 3, Ms. C starts to produce breastmilk and requests to breastfeed. At this time, the multidisciplinary team determines she is not able to visit her child in the NICU due to psychiatric instability. No plan is developed to facilitate hand expression or pumping of breastmilk while Ms. C is on the psychiatric unit. The clinical teams discuss whether the benefits of breastfeeding and/or pumping breastmilk would outweigh the risks. CPS determines that Ms. C is unable to retain custody and places the child in kinship foster care while awaiting clinical improvement from her.

CASE 2

Ms. S, age 32, has a history of schizophrenia. She lives with her husband and parents. She is pregnant for the first time and has been receiving consistent prenatal care. Ms. S is brought to the hospital by her husband for bizarre behavior and paranoia after self-discontinuing risperidone 2 mg twice daily due to concern about the medication’s influence on her pregnancy. An ultrasound confirms she is 37 weeks pregnant. Psychiatry is consulted because Ms. S is internally preoccupied, delusional, and endorses auditory hallucinations. She requires involuntary hospitalization for decompensated psychosis. During admission, Ms. S experiences improvement of her psychiatric symptoms while receiving risperidone 2 mg twice daily, which she takes consistently after receiving extensive psychoeducation regarding its safety profile during pregnancy and lactation.

After 2 weeks on the psychiatry unit, Ms. S’s care team transfers her to the obstetrics service with one-to-one supervision. At 39 weeks gestation, she has a vaginal delivery without complications. Because there are no concerns about infant harm, obstetrics, pediatrics, and psychiatry coordinate care so the baby can room in with Ms. S, her husband, and a staff supervisor to facilitate bonding. Ms. S starts to lactate, wishes to breastfeed, and meets with lactation, pediatric, obstetric, and psychiatric specialists to discuss the risks and benefits of breastfeeding and pumping breastmilk. She pursues direct breastfeeding until the baby is discharged home with the husband at postpartum Day 2. CPS is not called because there are no concerns for parental abuse or neglect at the infant’s discharge.

On postpartum Day 2, the obstetrics service transfers Ms. S back to the psychiatric unit for further treatment of her paranoia. She wishes to pump breastmilk while hospitalized, so the treatment team supplies a breast pump, facilitates the storage of breastmilk, and coordinates supervision during pumping to reduce the ligature risk. Ms. S’s husband visits daily to transport the milk and feed the infant breastmilk and formula to meet its nutritional needs. Ms. S maintains psychiatric stability while breast pumping, and the team helps transition her to breastfeeding during visitation with her husband and infant until she is discharged home at 2 weeks postpartum.

Continue to: Approaching care with a relational ethics framework

 

 

Approaching care with a relational ethics framework

A relational ethics framework was constructed to evaluate whether to support breastfeeding for both patients during their psychiatric hospitalizations. A relational ethics perspective is defined as “a moral responsibility within a context of human relations” [that] “recognizes the human interdependency and reciprocity within which personal autonomy is embedded.”2 This framework values connectedness and commonality between various and even conflicting parties. In the setting of a clinician-patient relationship, health care decisions are made with consideration of the patient’s traditional beliefs, values, and principles rather than the application of impartial moral principles. For these complex cases, this framework was chosen to determine the safest possible outcome for both mother and child.

Risks/benefits of breastfeeding by patients who have SMI

There are several methods of breastfeeding, including direct breastfeeding and other ways of expressing breastmilk such as pumping or hand expression.3 Unlike other forms of feeding using breastmilk, direct breastfeeding has been extensively studied, has well-established medical and psychological benefits for newborns and mothers, and enhances long-term bonding.4 Compared with their counterparts who do not breastfeed, mothers who breastfeed have lower rates of unintended pregnancy, cardiovascular disease, postpartum bleeding, osteoporosis, and breast and ovarian cancer.5 Among its key psychological benefits, breastfeeding is associated with an increase in maternal self-efficacy and, in some research, has been shown to be associated with a decreased risk of postpartum depression and stress.Additionally, breastfed infants experience lower rates of childhood infection and obesity, and improved nutrition, cognitive development, and immune function.6 The American Academy of Pediatrics recognizes these benefits and recommends that women exclusively breastfeed for 6 months postpartum and continue to breastfeed for 2 years or beyond if mutually desired by the mother and child.7 Absolute contraindications to breastfeeding must be ruled out (eg, infant classic galactosemia; maternal use of illicit substances such as cocaine, opioids, or phencyclidine; maternal HIV infection, etc).

The risks of breastfeeding by patients who have SMI must also be considered. In severe situations, the infant can be exposed to a mother’s agitation secondary to psychosis.8,9 The transmission of antipsychotic medication through breastmilk and associated adverse effects (eg, sedation, poor feeding, and extrapyramidal symptoms) are also potential risks and varies among different antipsychotic medications.1,10 Therefore, when prescribing an antipsychotic for a patient with SMI who breastfeeds, it is crucial to consider the medication’s safety profile as well as other factors, such as the relative infant dose (the weight-adjusted [ie, mg/kg] percentage of the maternal dosage ingested by a fully breastfed infant) and the molecular characteristics of the medication.10-12 Neonates should be routinely monitored for adverse effects, medication toxicity, and withdrawal symptoms, and care should be coordinated with the infant’s pediatrician. Certain antipsychotic medications, such as aripiprazole, may impact breastmilk production through the dopamine agonist’s interference of the prolactin reflex and anticholinergic properties.11,13 For a patient with SMI, perhaps the most significant risk involves the time and resources needed for breastfeeding, which can interfere with sleep and psychiatric treatment and possibly further exacerbate psychiatric symptoms.14-16 Additionally, breastfeeding difficulties or disruption can increase the risk of psychiatric symptoms and psychological distress.17 In Ms. C’s case, there was a delay in the baby latching as well as multiple medical and psychiatric factors that hindered the milk-ejection reflex to properly initiate; both of these factors rendered breastfeeding particularly difficult while Ms. C was on the inpatient psychiatry unit.17 In comparison, Ms. S was able to bond with her infant shortly after delivery, which facilitated the milk-ejection reflex and lactation.

Patients who wish to directly breastfeed but struggle to do so while tending to their acute psychiatric condition can benefit from expression of breastmilk that can be provided to the infant or discarded to facilitate breastfeeding in the future.18 While expression of breastmilk may not be as advantageous for infant health as direct breastfeeding due to the potential changes in breastmilk composition from collecting, storing, and heating, this option can be more protective than formula feeding and facilitate future breastfeeding.19 In these clinical scenarios, it is standard care to provide a hospital-grade breast pump to the patient, much like a continuous positive airway pressure machine is provided to patients with obstructive sleep apnea.20 However, there is often considerable difficulty obtaining proper breastfeeding equipment and a lack of services devoted to perinatal care in general inpatient settings. Barriers to direct breastfeeding and pumping of breastmilk are highlighted in the Table.21

CP02212e16_t1.png

Limitations on breastfeeding on an inpatient unit

The limitations in care and restrictions placed on breastfeeding are more optimally addressed in a mother and baby unit (MBU). MBUs are specialized inpatient psychiatric units designed for mothers experiencing severe perinatal psychiatric difficulties. Unlike general psychiatric units, MBUs allow for joint, full-time admission of mothers and their infants. These units also include multidisciplinary staff who specialize in treating perinatal mental health issues as well as infant care and child development.22 Admission into an MBU is considered best practice for new mothers requiring treatment, particularly in the United Kingdom, Australia, and France, as it is well-recognized that the separation of mother and baby can be psychologically harmful.23 In the UK, most patients admitted to an MBU showed significant improvement of their psychiatric symptoms and reported overall high satisfaction with care.24,25 Patients who experience postpartum psychosis prefer MBUs over general psychiatric units because the latter often lack specialized perinatal support, appropriate visitor arrangements, and adequate time with their infant.26-28

Continue to: The resistance to adopting MBUs in the United States...

 

 

The resistance to adopting MBUs in the United States has posed significant barriers in care for perinatal patients and has been attributed to financial barriers, medicolegal risk, staffing, and safety concerns.29 Though currently there are no MBUs in the US, other specialized units have been created. A partial day hospitalization program created in 2000 in Rhode Island for mothers and infants revolutionized the psychiatric care experience for new mothers.30 Since then, other institutions have significantly expanded their services to include perinatal psychiatry inpatient units, yet unlike MBUs, these units typically do not provide overnight rooming-in with infants.31 They have the necessary resources and facilities to accommodate the mother’s needs and maximize positive mother-infant interaction, while actively integrating the infant into the mother’s treatment. Breast pumping is treated as a necessary medical procedure and patients can easily access hospital-grade breast pumps with staff supervision. At one such perinatal psychiatric inpatient unit, high rates of treatment satisfaction and significant improvements in symptoms of depression, anxiety, active suicidal ideation, and overall functioning were observed at discharge.32 Therefore, it is crucial to incorporate strategies in general psychiatry units to improve perinatal care, acknowledging that most patients will not have access to these specialized units.21

A framework to approaching the relational ethics decisions

An interdisciplinary team used a relational ethics perspective to carefully analyze the risks and benefits of these complex cases. In Figure 1, we propose a framework for the relational ethics decisions of breastfeeding on general inpatient psychiatric units. In creating this framework, we considered principles of autonomy, beneficence, and nonmaleficence, along with the medical and logistical barriers to breastfeeding.

CP02212e16_f1.png

In Ms. C’s case, the team determined that the risks—which included disrupting the mother’s psychiatric treatment, exposing her to psychological harm due to increasing attachment before remanding the child to CPS custody, and risks to the child due to potential unpredictable agitation driven by the treatment-refractory psychosis of the mother as well as that of other psychiatric patients—outweighed the benefits of breastfeeding. We instead recommended breast pumping as an alternative once Ms. C’s psychiatric stability improved. We presented Ms. C with the option of breast pumping on postpartum Day 5. During a 1-day period in which she showed improved behavioral control, she was counseled on the risks and benefits of breastfeeding and exclusive pumping and was notified that the team would help her with the necessary resources, including consultation with a lactation specialist and breast pump. Despite lactation consultant support, Ms. C had low milk production and difficulty with hand expression, which was very discouraging to her. She produced 1 ounce of milk that was shared with the newborn while in the NICU. Because Ms. C’s psychiatric symptoms continued to be severe, with lability and aggression, and because pumping was triggering distress, the multidisciplinary team determined the best course of care would be to focus on her psychiatric recovery rather than on pumping breastmilk. To reduce milk production and minimize discomfort secondary to breast engorgement, the lactation consultant recommended cold compresses, pain management, and compression of breasts. Ultimately, the mother-infant dyad was unable to reap the benefits of breastfeeding (via pumping or direct breastfeeding) due to the mother’s underlying psychiatric illness, although the staffing, psychosocial support, and logistical limitations contributed to this outcome.33

In Ms. S’s case, the treatment team determined that there were no medical or psychiatric contraindications to breastfeeding, and she was counseled on the risks and benefits of direct breastfeeding and pumping. The treatment team determined it was safe for Ms. S to directly breastfeed as there were no concerns for infant harm post­delivery with constant supervision while on the obstetrics floor. The patient opted to directly breastfeed, which was successful with the guidance of a lactation specialist. When she was transferred to the psychiatric unit on postpartum Day 2, her child was discharged home with the husband. The patient was then encouraged to pump while the psychiatrists monitored her symptoms closely and facilitated increased staff and resources. Transportation of breastmilk was made possible by the family, and on postpartum Day 5, as the patient maintained psychiatric stability, the team discussed with Ms. S and her husband the prospect of direct breastfeeding. The treatment team arranged for separate visitation hours to minimize the possibility of exposing the infant to aggression from other patients on the unit and advocated with hospital leadership to approve of infant visitation on the unit.

Impact of involvement of Child Protective Services

The involvement of CPS also added complexity to Ms. C’s case. Without proper legal guidance, mothers with psychosis who lose custody can find it difficult to navigate the legal system and maintain contact with their children.34 As the prevalence of custody loss in mothers with psychosis is high (approximately 50% according to research published in the last 10 years), effective interventions to reunite the mother and child must be promoted (Figure 2).35-39 Ultimately, the goal of psychiatric hospitalization for perinatal women who have SMI is psychiatric stabilization. The preemptive involvement of psychiatry is crucial because it can allow for early postpartum planning and can provide an opportunity to address feeding options and custody concerns with the patient, social supports and services, and various medical teams. In Ms. C’s case, she visited her baby in the NICU on postpartum Day 2 without consultation with psychiatry or CPS, which posed risks to the patient, infant, and staff. It is vital that various clinicians collaborate with each other and the patient, working towards the goal of optimizing the patient’s mental health to allow for parenting rights in the future and maximizing a sustainable attachment between the parent and child. In Ms. S’s case, the husband was able to facilitate caring for the baby while the mother was hospitalized and played an integral role in the feeding process via pumped breastmilk and transport of the infant for direct breastfeeding.

CP02212e16_f2.png

Continue to: The differences in these 2 cases...

 

 

The differences in these 2 cases show the extreme importance of social support to benefit both the mother and child, and the need for more comprehensive social services for women who do not have a social safety net.

Bottom Line

These complex cases highlight an ethical decision-making approach to breastfeeding in perinatal women who have serious mental illness. Collaborative care and shared decision-making, which highlight the interests of the mother and baby, are crucial when assessing the risks and benefits of breastfeeding and pumping breastmilk. Our relational ethics framework can be used to better evaluate and implement breastfeeding options on general psychiatric units.

Related Resources

Drug Brand Names

Aripiprazole • Abilify
Olanzapine • Zyprexa
Risperidone • Risperdal

[embed:render:related:node:240620]

[embed:render:related:node:262580]

References

1. Brunner E, Falk DM, Jones M, et al. Olanzapine in pregnancy and breastfeeding: a review of data from global safety surveillance. BMC Pharmacol Toxicol. 2013;14:38. doi:10.1186/2050-6511-14-38

2. Seeman MV. Relational ethics: when mothers suffer from psychosis. Arch Womens Ment Health. 2004;7(3):201-210. doi:10.1007/s00737-004-0054-8

3. Motee A, Jeewon R. Importance of exclusive breastfeeding and complementary feeding among infants. Curr Res Nutr Food Sci. 2014;2(2). doi:10.12944/CRNFSJ.2.2.02

4. Committee Opinion No. 570: breastfeeding in underserved women: increasing initiation and continuation of breastfeeding. Obstet Gynecol. 2013;122(2 Pt 1):423-427. doi:10.1097/01.AOG.0000433008.93971.6a

5. Sibolboro Mezzacappa E, Endicott J. Parity mediates the association between infant feeding method and maternal depressive symptoms in the postpartum. Arch Womens Ment Health. 2007;10(6):259-266. doi:10.1007/s00737-007-0207-7

6. Kramer MS, Chalmers B, Hodnett ED, et al. Promotion of Breastfeeding Intervention Trial (PROBIT): a randomized trial in the Republic of Belarus. JAMA. 2001;285(4):413-420. doi:10.1001/jama.285.4.413

7. American Academy of Pediatrics. American Academy of Pediatrics calls for more support for breastfeeding mothers within updated policy recommendations. June 27, 2022. Accessed October 4, 2022. https://www.aap.org/en/news-room/news-releases/aap/2022/american-academy-of-pediatrics-calls-for-more-support-for-breastfeeding-mothers-within-updated-policy-recommendations

8. Hipwell AE, Kumar R. Maternal psychopathology and prediction of outcome based on mother-infant interaction ratings (BMIS). Br J Psychiatry. 1996;169(5):655-661. doi:10.1192/bjp.169.5.655

9. Chandra PS, Bhargavaraman RP, Raghunandan VN, et al. Delusions related to infant and their association with mother-infant interactions in postpartum psychotic disorders. Arch Womens Ment Health. 2006;9(5):285-288. doi:10.1007/s00737-006-0147-7

10. Klinger G, Stahl B, Fusar-Poli P, et al. Antipsychotic drugs and breastfeeding. Pediatr Endocrinol Rev. 2013;10(3):308-317.

11. Uguz F. A new safety scoring system for the use of psychotropic drugs during lactation. Am J Ther. 2021;28(1):e118-e126. doi:10.1097/MJT.0000000000000909

12. Hale TW, Krutsch K. Hale’s Medications & Mothers’ Milk, 2023: A Manual of Lactational Pharmacology. 20th ed. Springer Publishing Company; 2023.

13. Komaroff A. Aripiprazole and lactation failure: the importance of shared decision making. A case report. Case Rep Womens Health. 2021;30:e00308. doi:10.1016/j.crwh.2021.e00308‌

14. Dennis CL, McQueen K. Does maternal postpartum depressive symptomatology influence infant feeding outcomes? Acta Pediatr. 2007;96(4):590-594. doi:10.1111/j.1651-2227.2007.00184.x

15. Chaput KH, Nettel-Aguirre A, Musto R, et al. Breastfeeding difficulties and supports and risk of postpartum depression in a cohort of women who have given birth in Calgary: a prospective cohort study. CMAJ Open. 2016;4(1):E103-E109. doi:10.9778/cmajo.20150009

16. Dias CC, Figueiredo B. Breastfeeding and depression: a systematic review of the literature. J Affect Disord. 2015;171:142-154. doi:10.1016/j.jad.2014.09.022

17. Brown A, Rance J, Bennett P. Understanding the relationship between breastfeeding and postnatal depression: the role of pain and physical difficulties. J Adv Nurs. 2016;72(2):273-282. doi:10.1111/jan.12832

18. Rosenbaum KA. Exclusive breastmilk pumping: a concept analysis. Nurs Forum. 2022;57(5):946-953. doi:10.1111/nuf.12766

19. Boone KM, Geraghty SR, Keim SA. Feeding at the breast and expressed milk feeding: associations with otitis media and diarrhea in infants. J Pediatr. 2016;174:118-125. doi:10.1016/j.jpeds.2016.04.006

20. Epstein LJ, Kristo D, Strollo PJ Jr, et al; Adult Obstructive Sleep Apnea Task Force of the American Academy of Sleep Medicine. Clinical guideline for the evaluation, management and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med. 2009;5(3):263-276.

21. Caan MP, Sreshta NE, Okwerekwu JA, et al. Clinical and legal considerations regarding breastfeeding on psychiatric units. J Am Acad Psychiatry Law. 2022;50(2):200-207. doi:10.29158/JAAPL.210086-21

22. Glangeaud-Freudenthal NMC, Rainelli C, Cazas O, et al. Inpatient mother and baby psychiatric units (MBUs) and day cares. In: Sutter-Dallay AL, Glangeaud-Freudenthal NC, Guedeney A, et al, eds. Joint Care of Parents and Infants in Perinatal Psychiatry. Springer, Cham; 2016:147-164. doi:10.1007/978-3-319-21557-0_10

23. Dembosky A. A humane approach to caring for new mothers in psychiatric crisis. Health Aff (Millwood). 2021;40(10):1528-1533. doi:10.1377/hlthaff.2021.01288

24. Connellan K, Bartholomaeus C, Due C, et al. A systematic review of research on psychiatric mother-baby units. Arch Womens Ment Health. 2017;20(3):373-388. doi:10.1007/s00737-017-0718-9

25. Griffiths J, Lever Taylor B, Morant N, et al. A qualitative comparison of experiences of specialist mother and baby units versus general psychiatric wards. BMC Psychiatry. 2019;19(1):401. doi:10.1186/s12888-019-2389-8

26. Heron J, Gilbert N, Dolman C, et al. Information and support needs during recovery from postpartum psychosis. Arch Womens Ment Health. 2012;15(3):155-165. doi:10.1007/s00737-012-0267-1

27. Robertson E, Lyons A. Living with puerperal psychosis: a qualitative analysis. Psychol Psychother. 2003;76(Pt 4):411-431. doi:10.1348/147608303770584755

28. Mental Welfare Commission for Scotland. Perinatal Themed Visit Report: Keeping Mothers and Babies in Mind. Mental Welfare Commission for Scotland; 2016.

29. Wisner KL, Jennings KD, Conley B. Clinical dilemmas due to the lack of inpatient mother-baby units. Int J Psychiatry Med. 1996;26(4):479-493. doi:10.2190/NFJK-A4V7-CXUU-AM89

30. Battle CL, Howard MM. A mother-baby psychiatric day hospital: history, rationale, and why perinatal mental health is important for obstetric medicine. Obstet Med. 2014;7(2):66-70. doi:10.1177/1753495X13514402

31. Bullard ES, Meltzer-Brody S, Rubinow DR. The need for comprehensive psychiatric perinatal care-the University of North Carolina at Chapel Hill, Department of Psychiatry, Center for Women’s Mood Disorders launches the first dedicated inpatient program in the United States. Am J Obstet Gynecol. 2009;201(5):e10-e11. doi:10.1016/j.ajog.2009.05.004

32. Meltzer-Brody S, Brandon AR, Pearson B, et al. Evaluating the clinical effectiveness of a specialized perinatal psychiatry inpatient unit. Arch Womens Ment Health. 2014;17(2):107-113. doi:10.1007/s00737-013-0390-7

33. Alvarez-Toro V. Gender-specific care for women in psychiatric units. J Am Acad Psychiatry Law. 2022;JAAPL.220015-21. doi:10.29158/JAAPL.220015-21

34. Diaz-Caneja A, Johnson S. The views and experiences of severely mentally ill mothers--a qualitative study. Soc Psychiatry Psychiatr Epidemiol. 2004;39(6):472-482. doi:10.1007/s00127-004-0772-2

35. Gewurtz R, Krupa T, Eastabrook S, et al. Prevalence and characteristics of parenting among people served by assertive community treatment. Psychiatr Rehabil J. 2004;28(1):63-65. doi:10.2975/28.2004.63.65

36. Howard LM, Kumar R, Thornicroft G. Psychosocial characteristics and needs of mothers with psychotic disorders. Br J Psychiatry. 2001;178:427-432. doi:10.1192/bjp.178.5.427

37. Hollingsworth LD. Child custody loss among women with persistent severe mental illness. Social Work Research. 2004;28(4):199-209. doi:10.1093/swr/28.4.199

38. Dipple H, Smith S, Andrews H, et al. The experience of motherhood in women with severe and enduring mental illness. Soc Psychiatry Psychiatr Epidemiolf. 2002;37(7):336-340. doi:10.1007/s00127-002-0559-2

39. Seeman MV. Intervention to prevent child custody loss in mothers with schizophrenia. Schizophr Res Treatment. 2012;2012:796763. doi:10.1155/2012/796763

References

1. Brunner E, Falk DM, Jones M, et al. Olanzapine in pregnancy and breastfeeding: a review of data from global safety surveillance. BMC Pharmacol Toxicol. 2013;14:38. doi:10.1186/2050-6511-14-38

2. Seeman MV. Relational ethics: when mothers suffer from psychosis. Arch Womens Ment Health. 2004;7(3):201-210. doi:10.1007/s00737-004-0054-8

3. Motee A, Jeewon R. Importance of exclusive breastfeeding and complementary feeding among infants. Curr Res Nutr Food Sci. 2014;2(2). doi:10.12944/CRNFSJ.2.2.02

4. Committee Opinion No. 570: breastfeeding in underserved women: increasing initiation and continuation of breastfeeding. Obstet Gynecol. 2013;122(2 Pt 1):423-427. doi:10.1097/01.AOG.0000433008.93971.6a

5. Sibolboro Mezzacappa E, Endicott J. Parity mediates the association between infant feeding method and maternal depressive symptoms in the postpartum. Arch Womens Ment Health. 2007;10(6):259-266. doi:10.1007/s00737-007-0207-7

6. Kramer MS, Chalmers B, Hodnett ED, et al. Promotion of Breastfeeding Intervention Trial (PROBIT): a randomized trial in the Republic of Belarus. JAMA. 2001;285(4):413-420. doi:10.1001/jama.285.4.413

7. American Academy of Pediatrics. American Academy of Pediatrics calls for more support for breastfeeding mothers within updated policy recommendations. June 27, 2022. Accessed October 4, 2022. https://www.aap.org/en/news-room/news-releases/aap/2022/american-academy-of-pediatrics-calls-for-more-support-for-breastfeeding-mothers-within-updated-policy-recommendations

8. Hipwell AE, Kumar R. Maternal psychopathology and prediction of outcome based on mother-infant interaction ratings (BMIS). Br J Psychiatry. 1996;169(5):655-661. doi:10.1192/bjp.169.5.655

9. Chandra PS, Bhargavaraman RP, Raghunandan VN, et al. Delusions related to infant and their association with mother-infant interactions in postpartum psychotic disorders. Arch Womens Ment Health. 2006;9(5):285-288. doi:10.1007/s00737-006-0147-7

10. Klinger G, Stahl B, Fusar-Poli P, et al. Antipsychotic drugs and breastfeeding. Pediatr Endocrinol Rev. 2013;10(3):308-317.

11. Uguz F. A new safety scoring system for the use of psychotropic drugs during lactation. Am J Ther. 2021;28(1):e118-e126. doi:10.1097/MJT.0000000000000909

12. Hale TW, Krutsch K. Hale’s Medications & Mothers’ Milk, 2023: A Manual of Lactational Pharmacology. 20th ed. Springer Publishing Company; 2023.

13. Komaroff A. Aripiprazole and lactation failure: the importance of shared decision making. A case report. Case Rep Womens Health. 2021;30:e00308. doi:10.1016/j.crwh.2021.e00308‌

14. Dennis CL, McQueen K. Does maternal postpartum depressive symptomatology influence infant feeding outcomes? Acta Pediatr. 2007;96(4):590-594. doi:10.1111/j.1651-2227.2007.00184.x

15. Chaput KH, Nettel-Aguirre A, Musto R, et al. Breastfeeding difficulties and supports and risk of postpartum depression in a cohort of women who have given birth in Calgary: a prospective cohort study. CMAJ Open. 2016;4(1):E103-E109. doi:10.9778/cmajo.20150009

16. Dias CC, Figueiredo B. Breastfeeding and depression: a systematic review of the literature. J Affect Disord. 2015;171:142-154. doi:10.1016/j.jad.2014.09.022

17. Brown A, Rance J, Bennett P. Understanding the relationship between breastfeeding and postnatal depression: the role of pain and physical difficulties. J Adv Nurs. 2016;72(2):273-282. doi:10.1111/jan.12832

18. Rosenbaum KA. Exclusive breastmilk pumping: a concept analysis. Nurs Forum. 2022;57(5):946-953. doi:10.1111/nuf.12766

19. Boone KM, Geraghty SR, Keim SA. Feeding at the breast and expressed milk feeding: associations with otitis media and diarrhea in infants. J Pediatr. 2016;174:118-125. doi:10.1016/j.jpeds.2016.04.006

20. Epstein LJ, Kristo D, Strollo PJ Jr, et al; Adult Obstructive Sleep Apnea Task Force of the American Academy of Sleep Medicine. Clinical guideline for the evaluation, management and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med. 2009;5(3):263-276.

21. Caan MP, Sreshta NE, Okwerekwu JA, et al. Clinical and legal considerations regarding breastfeeding on psychiatric units. J Am Acad Psychiatry Law. 2022;50(2):200-207. doi:10.29158/JAAPL.210086-21

22. Glangeaud-Freudenthal NMC, Rainelli C, Cazas O, et al. Inpatient mother and baby psychiatric units (MBUs) and day cares. In: Sutter-Dallay AL, Glangeaud-Freudenthal NC, Guedeney A, et al, eds. Joint Care of Parents and Infants in Perinatal Psychiatry. Springer, Cham; 2016:147-164. doi:10.1007/978-3-319-21557-0_10

23. Dembosky A. A humane approach to caring for new mothers in psychiatric crisis. Health Aff (Millwood). 2021;40(10):1528-1533. doi:10.1377/hlthaff.2021.01288

24. Connellan K, Bartholomaeus C, Due C, et al. A systematic review of research on psychiatric mother-baby units. Arch Womens Ment Health. 2017;20(3):373-388. doi:10.1007/s00737-017-0718-9

25. Griffiths J, Lever Taylor B, Morant N, et al. A qualitative comparison of experiences of specialist mother and baby units versus general psychiatric wards. BMC Psychiatry. 2019;19(1):401. doi:10.1186/s12888-019-2389-8

26. Heron J, Gilbert N, Dolman C, et al. Information and support needs during recovery from postpartum psychosis. Arch Womens Ment Health. 2012;15(3):155-165. doi:10.1007/s00737-012-0267-1

27. Robertson E, Lyons A. Living with puerperal psychosis: a qualitative analysis. Psychol Psychother. 2003;76(Pt 4):411-431. doi:10.1348/147608303770584755

28. Mental Welfare Commission for Scotland. Perinatal Themed Visit Report: Keeping Mothers and Babies in Mind. Mental Welfare Commission for Scotland; 2016.

29. Wisner KL, Jennings KD, Conley B. Clinical dilemmas due to the lack of inpatient mother-baby units. Int J Psychiatry Med. 1996;26(4):479-493. doi:10.2190/NFJK-A4V7-CXUU-AM89

30. Battle CL, Howard MM. A mother-baby psychiatric day hospital: history, rationale, and why perinatal mental health is important for obstetric medicine. Obstet Med. 2014;7(2):66-70. doi:10.1177/1753495X13514402

31. Bullard ES, Meltzer-Brody S, Rubinow DR. The need for comprehensive psychiatric perinatal care-the University of North Carolina at Chapel Hill, Department of Psychiatry, Center for Women’s Mood Disorders launches the first dedicated inpatient program in the United States. Am J Obstet Gynecol. 2009;201(5):e10-e11. doi:10.1016/j.ajog.2009.05.004

32. Meltzer-Brody S, Brandon AR, Pearson B, et al. Evaluating the clinical effectiveness of a specialized perinatal psychiatry inpatient unit. Arch Womens Ment Health. 2014;17(2):107-113. doi:10.1007/s00737-013-0390-7

33. Alvarez-Toro V. Gender-specific care for women in psychiatric units. J Am Acad Psychiatry Law. 2022;JAAPL.220015-21. doi:10.29158/JAAPL.220015-21

34. Diaz-Caneja A, Johnson S. The views and experiences of severely mentally ill mothers--a qualitative study. Soc Psychiatry Psychiatr Epidemiol. 2004;39(6):472-482. doi:10.1007/s00127-004-0772-2

35. Gewurtz R, Krupa T, Eastabrook S, et al. Prevalence and characteristics of parenting among people served by assertive community treatment. Psychiatr Rehabil J. 2004;28(1):63-65. doi:10.2975/28.2004.63.65

36. Howard LM, Kumar R, Thornicroft G. Psychosocial characteristics and needs of mothers with psychotic disorders. Br J Psychiatry. 2001;178:427-432. doi:10.1192/bjp.178.5.427

37. Hollingsworth LD. Child custody loss among women with persistent severe mental illness. Social Work Research. 2004;28(4):199-209. doi:10.1093/swr/28.4.199

38. Dipple H, Smith S, Andrews H, et al. The experience of motherhood in women with severe and enduring mental illness. Soc Psychiatry Psychiatr Epidemiolf. 2002;37(7):336-340. doi:10.1007/s00127-002-0559-2

39. Seeman MV. Intervention to prevent child custody loss in mothers with schizophrenia. Schizophr Res Treatment. 2012;2012:796763. doi:10.1155/2012/796763

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
e16-e23
Page Number
e16-e23
Publications
Publications
Topics
Article Type
Display Headline
Breastfeeding by patients with serious mental illness: An ethical approach
Display Headline
Breastfeeding by patients with serious mental illness: An ethical approach
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1223 Soundararajan breastfeeding</fileName> <TBEID>0C02EE3F.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EE3F</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Breastfeeding by patients with s</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231212T065414</firstPublished> <LastPublished>20231212T065414</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231212T065414</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Sitara Soundararajan, MD, Marlee Madora, MD, Rubiahna Vaughn, MD</byline> <bylineText/> <bylineFull>Sitara Soundararajan, MD, Marlee Madora, MD, Rubiahna Vaughn, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>e16-e23</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Difficult ethical situations can arise when treating perinatal women who have serious mental illness (SMI). Clinicians must consider ethical issues related to a</metaDescription> <articlePDF>299600</articlePDF> <teaserImage/> <title>Breastfeeding by patients with serious mental illness: An ethical approach Our framework can guide decisions about breastfeeding on a psychiatric unit</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords> <keyword>schizophrenia</keyword> <keyword> bipolar disorder</keyword> <keyword> depression</keyword> <keyword> breastfeeding</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">26935</term> </sections> <topics> <term canonical="true">293</term> <term>190</term> <term>202</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/1800267c.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Breastfeeding by patients with serious mental illness: An ethical approach Our framework can guide decisions about breastfeeding on a psychiatric unit</title> <deck/> </itemMeta> <itemContent> <p>Difficult ethical situations can arise when treating perinatal women who have serious mental illness (SMI). Clinicians must consider ethical issues related to administering antipsychotic medications, the safety of breastfeeding, and concerns for child welfare. They need to carefully weigh the risks and benefits of each decision when treating perinatal women who have SMI. Ethical guidelines can help clinicians best support families in these situations.</p> <p>In this article, we describe 2 cases of women with psychotic disorders who requested to breastfeed after delivering their child during an inpatient psychiatric hospitalization. The course of their hospitalizations illustrated common ethical questions and facilitated the creation of a framework to assist with complex decision-making regarding breastfeeding on inpatient psychiatric units.</p> <h3> <span class="dingbat"> </span> <span class="box">CASE 1</span> <span class="dingbat"> </span> </h3> <p>Ms. C, age 41, is multigravida with a psychiatric history of chronic, severe schizoaffective disorder and lives in supportive housing. When Ms. C presents to the hospital in search of a rape kit, clinicians discover she is 22 weeks pregnant but has not received any prenatal care. Psychiatry is consulted because she is found to be intermittently agitated and endorses grandiose delusions. Ms. C requires involuntary hospitalization for decompensated psychosis because she refuses prenatal and psychiatric care. Because it has reassuring reproductive safety data,<sup>1</sup> olanzapine 5 mg/d is started. However, Ms. C experiences minimal improvement from a maximum dose of 20 mg/d. After 13 weeks on the psychiatry unit, she is transferred to obstetrics service for preeclampsia with severe features. Ms. C requires an urgent cesarean delivery at 37 weeks. Her baby boy is transferred to the neonatal intensive care unit (NICU) for transient tachypnea. After delivery and in consultation with psychiatry, the pediatrics team calls Child Protective Services (CPS) due to concern for neglect driven by Ms. C’s psychiatric condition. Ms. C visits the child with medical unit staff supervision in the NICU without consulting with the psychiatry service or CPS. On postpartum Day 2, Ms. C is transferred back to psychiatry for persistent psychosis.</p> <p>On postpartum Day 3, Ms. C starts to produce breastmilk and requests to breastfeed. At this time, the multidisciplinary team determines she is not able to visit her child in the NICU due to psychiatric instability. No plan is developed to facilitate hand expression or pumping of breastmilk while Ms. C is on the psychiatric unit. The clinical teams discuss whether the benefits of breastfeeding and/or pumping breastmilk would outweigh the risks. CPS determines that Ms. C is unable to retain custody and places the child in kinship foster care while awaiting clinical improvement from her.</p> <h3> <span class="dingbat"> </span> <span class="box">CASE 2</span> <span class="dingbat"> </span> </h3> <p>Ms. S, age 32, has a history of schizophrenia. She lives with her husband and parents. She is pregnant for the first time and has been receiving consistent prenatal care. Ms. S is brought to the hospital by her husband for bizarre behavior and paranoia after self-discontinuing risperidone 2 mg twice daily due to concern about the medication’s influence on her pregnancy. An ultrasound confirms she is 37 weeks pregnant. Psychiatry is consulted because Ms. S is internally preoccupied, delusional, and endorses auditory hallucinations. She requires involuntary hospitalization for decompensated psychosis. During admission, Ms. S experiences improvement of her psychiatric symptoms while receiving risperidone 2 mg twice daily, which she takes consistently after receiving extensive psychoeducation regarding its safety profile during pregnancy and lactation.</p> <p>After 2 weeks on the psychiatry unit, Ms. S’s care team transfers her to the obstetrics service with one-to-one supervision. At 39 weeks gestation, she has a vaginal delivery without complications. Because there are no concerns about infant harm, obstetrics, pediatrics, and psychiatry coordinate care so the baby can room in with Ms. S, her husband, and a staff supervisor to facilitate bonding. Ms. S starts to lactate, wishes to breastfeed, and meets with lactation, pediatric, obstetric, and psychiatric specialists to discuss the risks and benefits of breastfeeding and pumping breastmilk. She pursues direct breastfeeding until the baby is discharged home with the husband at postpartum Day 2. CPS is not called because there are no concerns for parental abuse or neglect at the infant’s discharge.<br/><br/>On postpartum Day 2, the obstetrics service transfers Ms. S back to the psychiatric unit for further treatment of her paranoia. She wishes to pump breastmilk while hospitalized, so the treatment team supplies a breast pump, facilitates the storage of breastmilk, and coordinates supervision during pumping to reduce the ligature risk. Ms. S’s husband visits daily to transport the milk and feed the infant breastmilk and formula to meet its nutritional needs. Ms. S maintains psychiatric stability while breast pumping, and the team helps transition her to breastfeeding during visitation with her husband and infant until she is discharged home at 2 weeks postpartum.</p> <h3>Approaching care with a relational ethics framework</h3> <p>A relational ethics framework was constructed to evaluate whether to support breastfeeding for both patients during their psychiatric hospitalizations. A relational ethics perspective is defined as “a moral responsibility within a context of human relations” [that] “recognizes the human interdependency and reciprocity within which personal autonomy is embedded.”<sup>2</sup> This framework values connectedness and commonality between various and even conflicting parties. In the setting of a clinician-patient relationship, health care decisions are made with consideration of the patient’s traditional beliefs, values, and principles rather than the application of impartial moral principles. For these complex cases, this framework was chosen to determine the safest possible outcome for both mother and child.</p> <h3>Risks/benefits of breastfeeding by patients who have SMI</h3> <p>There are several methods of breastfeeding, including direct breastfeeding and other ways of expressing breastmilk such as pumping or hand expression.<sup>3</sup> Unlike other forms of feeding using breastmilk, direct breastfeeding has been extensively studied, has well-established medical and psychological benefits for newborns and mothers, and enhances long-term bonding.<sup>4</sup> Compared with their counterparts who do not breastfeed, mothers who breastfeed have lower rates of unintended pregnancy, cardiovascular disease, postpartum bleeding, osteoporosis, and breast and ovarian cancer.<sup>5 </sup>Among its key psychological benefits, breastfeeding is associated with an increase in maternal self-efficacy and, in some research, has been shown to be associated with a decreased risk of postpartum depression and stress.<sup> </sup>Additionally, breastfed infants experience lower rates of childhood infection and obesity, and improved nutrition, cognitive development, and immune function.<sup>6</sup> The American Academy of Pediatrics recognizes these benefits and recommends that women exclusively breastfeed for 6 months postpartum and continue to breastfeed for 2 years or beyond if mutually desired by the mother and child.<sup>7</sup> Absolute contraindications to breastfeeding must be ruled out (eg, infant classic galactosemia; maternal use of illicit substances such as cocaine, opioids, or phencyclidine; maternal HIV infection, etc).</p> <p>The risks of breastfeeding by patients who have SMI must also be considered. In severe situations, the infant can be exposed to a mother’s agitation secondary to psychosis.<sup>8,9</sup> The transmission of antipsychotic medication through breastmilk and associated adverse effects (eg, sedation, poor feeding, and extrapyramidal symptoms) are also potential risks and varies among different antipsychotic medications.<sup>1,10 </sup>Therefore, when prescribing an antipsychotic for a patient with SMI who breastfeeds, it is crucial to consider the medication’s safety profile as well as other factors, such as the relative infant dose (the weight-adjusted [ie, mg/kg] percentage of the maternal dosage ingested by a fully breastfed infant) and the molecular characteristics of the medication.<sup>10-12</sup> Neonates should be routinely monitored for adverse effects, medication toxicity, and withdrawal symptoms, and care should be coordinated with the infant’s pediatrician. Certain antipsychotic medications, such as aripiprazole, may impact breastmilk production through the dopamine agonist’s interference of the prolactin reflex and anticholinergic properties.<sup>11,13</sup> For a patient with SMI, perhaps the most significant risk involves the time and resources needed for breastfeeding, which can interfere with sleep and psychiatric treatment and possibly further exacerbate psychiatric symptoms.<sup>14-16</sup> Additionally, breastfeeding difficulties or disruption can increase the risk of psychiatric symptoms and psychological distress.<sup>17 </sup>In Ms. C’s case, there was a delay in the baby latching as well as multiple medical and psychiatric factors that hindered the milk-ejection reflex to properly initiate; both of these factors rendered breastfeeding particularly difficult while Ms. C was on the inpatient psychiatry unit.<sup>17</sup> In comparison, Ms. S was able to bond with her infant shortly after delivery, which facilitated the milk-ejection reflex and lactation.<br/><br/>Patients who wish to directly breastfeed but struggle to do so while tending to their acute psychiatric condition can benefit from expression of breastmilk that can be provided to the infant or discarded to facilitate breastfeeding in the future.<sup>18 </sup>While expression of breastmilk may not be as advantageous for infant health as direct breastfeeding due to the potential changes in breastmilk composition from collecting, storing, and heating, this option can be more protective than formula feeding and facilitate future breastfeeding.<sup>19 </sup>In these clinical scenarios, it is standard care to provide a hospital-grade breast pump to the patient, much like a continuous positive airway pressure machine is provided to patients with obstructive sleep apnea.<sup>20</sup> However, there is often considerable difficulty obtaining proper breastfeeding equipment and a lack of services devoted to perinatal care in general inpatient settings. Barriers to direct breastfeeding and pumping of breastmilk are highlighted in the <span class="bitalic">Table</span>.<sup>21</sup></p> <h3>Limitations on breastfeeding on an inpatient unit</h3> <p>The limitations in care and restrictions placed on breastfeeding are more optimally addressed in a mother and baby unit (MBU). MBUs are specialized inpatient psychiatric units designed for mothers experiencing severe perinatal psychiatric difficulties. Unlike general psychiatric units, MBUs allow for joint, full-time admission of mothers and their infants. These units also include multidisciplinary staff who specialize in treating perinatal mental health issues as well as infant care and child development.<sup>22</sup> Admission into an MBU is considered best practice for new mothers requiring treatment, particularly in the United Kingdom, Australia, and France, as it is well-recognized that the separation of mother and baby can be psychologically harmful.<sup>23</sup> In the UK, most patients admitted to an MBU showed significant improvement of their psychiatric symptoms and reported overall high satisfaction with care.<sup>24,25</sup> Patients who experience postpartum psychosis prefer MBUs over general psychiatric units because the latter often lack specialized perinatal support, appropriate visitor arrangements, and adequate time with their infant.<sup>26-28</sup></p> <p>The resistance to adopting MBUs in the United States has posed significant barriers in care for perinatal patients and has been attributed to financial barriers, medicolegal risk, staffing, and safety concerns.<sup>29</sup> Though currently there are no MBUs in the US, other specialized units have been created. A partial day hospitalization program created in 2000 in Rhode Island for mothers and infants revolutionized the psychiatric care experience for new mothers.<sup>30</sup> Since then, other institutions have significantly expanded their services to include perinatal psychiatry inpatient units, yet unlike MBUs, these units typically do not provide overnight rooming-in with infants.<sup>31</sup> They have the necessary resources and facilities to accommodate the mother’s needs and maximize positive mother-infant interaction, while actively integrating the infant into the mother’s treatment. Breast pumping is treated as a necessary medical procedure and patients can easily access hospital-grade breast pumps with staff supervision. At one such perinatal psychiatric inpatient unit, high rates of treatment satisfaction and significant improvements in symptoms of depression, anxiety, active suicidal ideation, and overall functioning were observed at discharge.<sup>32</sup> Therefore, it is crucial to incorporate strategies in general psychiatry units to improve perinatal care, acknowledging that most patients will not have access to these specialized units.<sup>21</sup></p> <h3>A framework to approaching the relational ethics decisions</h3> <p>An interdisciplinary team used a relational ethics perspective to carefully analyze the risks and benefits of these complex cases. In <b><i>Figure 1</i></b>, we propose a framework for the relational ethics decisions of breastfeeding on general inpatient psychiatric units. In creating this framework, we considered principles of autonomy, beneficence, and nonmaleficence, along with the medical and logistical barriers to breastfeeding. </p> <p>In Ms. C’s case, the team determined that the risks—which included disrupting the mother’s psychiatric treatment, exposing her to psychological harm due to increasing attachment before remanding the child to CPS custody, and risks to the child due to potential unpredictable agitation driven by the treatment-refractory psychosis of the mother as well as that of other psychiatric patients—outweighed the benefits of breastfeeding. We instead recommended breast pumping as an alternative once Ms. C’s psychiatric stability improved. We presented Ms. C with the option of breast pumping on postpartum Day 5. During a 1-day period in which she showed improved behavioral control, she was counseled on the risks and benefits of breastfeeding and exclusive pumping and was notified that the team would help her with the necessary resources, including consultation with a lactation specialist and breast pump. Despite lactation consultant support, Ms. C had low milk production and difficulty with hand expression, which was very discouraging to her. She produced 1 ounce of milk that was shared with the newborn while in the NICU. Because Ms. C’s psychiatric symptoms continued to be severe, with lability and aggression, and because pumping was triggering distress, the multidisciplinary team determined the best course of care would be to focus on her psychiatric recovery rather than on pumping breastmilk. To reduce milk production and minimize discomfort secondary to breast engorgement, the lactation consultant recommended cold compresses, pain management, and compression of breasts. Ultimately, the mother-infant dyad was unable to reap the benefits of breastfeeding (via pumping or direct breastfeeding) due to the mother’s underlying psychiatric illness, although the staffing, psychosocial support, and logistical limitations contributed to this outcome.<sup>33<br/><br/></sup>In Ms. S’s case, the treatment team determined that there were no medical or psychiatric contraindications to breastfeeding, and she was counseled on the risks and benefits of direct breastfeeding and pumping. The treatment team determined it was safe for Ms. S to directly breastfeed as there were no concerns for infant harm post­delivery with constant supervision while on the obstetrics floor. The patient opted to directly breastfeed, which was successful with the guidance of a lactation specialist. When she was transferred to the psychiatric unit on postpartum Day 2, her child was discharged home with the husband. The patient was then encouraged to pump while the psychiatrists monitored her symptoms closely and facilitated increased staff and resources. Transportation of breastmilk was made possible by the family, and on postpartum Day 5, as the patient maintained psychiatric stability, the team discussed with Ms. S and her husband the prospect of direct breastfeeding. The treatment team arranged for separate visitation hours to minimize the possibility of exposing the infant to aggression from other patients on the unit and advocated with hospital leadership to approve of infant visitation on the unit.</p> <h3>Impact of involvement of Child Protective Services</h3> <p>The involvement of CPS also added complexity to Ms. C’s case. Without proper legal guidance, mothers with psychosis who lose custody can find it difficult to navigate the legal system and maintain contact with their children.<sup>34</sup> As the prevalence of custody loss in mothers with psychosis is high (approximately 50% according to research published in the last 10 years), effective interventions to reunite the mother and child must be promoted (<span class="bitalic">Figure 2</span>, <span class="bitalic">page e21</span>).<sup>35-39</sup> Ultimately, the goal of psychiatric hospitalization for perinatal women who have SMI is psychiatric stabilization. The preemptive involvement of psychiatry is crucial because it can allow for early postpartum planning and can provide an opportunity to address feeding options and custody concerns with the patient, social supports and services, and various medical teams. In Ms. C’s case, she visited her baby in the NICU on postpartum Day 2 without consultation with psychiatry or CPS, which posed risks to the patient, infant, and staff. It is vital that various clinicians collaborate with each other and the patient, working towards the goal of optimizing the patient’s mental health to allow for parenting rights in the future and maximizing a sustainable attachment between the parent and child. In Ms. S’s case, the husband was able to facilitate caring for the baby while the mother was hospitalized and played an integral role in the feeding process via pumped breastmilk and transport of the infant for direct breastfeeding.</p> <p>The differences in these 2 cases show the extreme importance of social support to benefit both the mother and child, and the need for more comprehensive social services for women who do not have a social safety net.</p> <p> <strong>References</strong> </p> <p class="reference"> 1. Brunner E, Falk DM, Jones M, et al. Olanzapine in pregnancy and breastfeeding: a review of data from global safety surveillance. BMC Pharmacol Toxicol. 2013;14:38. doi:10.1186/2050-6511-14-38<br/><br/> 2. Seeman MV. Relational ethics: when mothers suffer from psychosis. Arch Womens Ment Health. 2004;7(3):201-210. doi:10.1007/s00737-004-0054-8<br/><br/> 3. Motee A, Jeewon R. Importance of exclusive breastfeeding and complementary feeding among infants. Curr Res Nutr Food Sci. 2014;2(2). doi:10.12944/CRNFSJ.2.2.02<br/><br/> 4. Committee Opinion No. 570: breastfeeding in underserved women: increasing initiation and continuation of breastfeeding. Obstet Gynecol. 2013;122(2 Pt 1):423-427. doi:10.1097/01.AOG.0000433008.93971.6a <br/><br/> 5. Sibolboro Mezzacappa E, Endicott J. Parity mediates the association between infant feeding method and maternal depressive symptoms in the postpartum. Arch Womens Ment Health. 2007;10(6):259-266. doi:10.1007/s00737-007-0207-7 <br/><br/> 6. Kramer MS, Chalmers B, Hodnett ED, et al. Promotion of Breastfeeding Intervention Trial (PROBIT): a randomized trial in the Republic of Belarus. JAMA. 2001;285(4):413-420. doi:10.1001/jama.285.4.413<br/><br/> 7. American Academy of Pediatrics. American Academy of Pediatrics calls for more support for breastfeeding mothers within updated policy recommendations. June 27, 2022. Accessed October 4, 2022. https://www.aap.org/en/news-room/news-releases/aap/2022/american-academy-of-pediatrics-calls-for-more-support-for-breastfeeding-mothers-within-updated-policy-recommendations <br/><br/> 8. Hipwell AE, Kumar R. Maternal psychopathology and prediction of outcome based on mother-infant interaction ratings (BMIS). Br J Psychiatry. 1996;169(5):655-661. doi:10.1192/bjp.169.5.655<br/><br/> 9. Chandra PS, Bhargavaraman RP, Raghunandan VN, et al. Delusions related to infant and their association with mother-infant interactions in postpartum psychotic disorders. Arch Womens Ment Health. 2006;9(5):285-288. doi:10.1007/s00737-006-0147-7<br/><br/> 10. Klinger G, Stahl B, Fusar-Poli P, et al. Antipsychotic drugs and breastfeeding. Pediatr Endocrinol Rev. 2013;10(3):308-317. <br/><br/> 11. Uguz F. A new safety scoring system for the use of psychotropic drugs during lactation. Am J Ther. 2021;28(1):e118-e126. doi:10.1097/MJT.0000000000000909<br/><br/> 12. Hale TW, Krutsch K. <i>Hale’s Medications &amp; Mothers’ Milk, 2023: A Manual of Lactational Pharmacology</i>. 20th ed. Springer Publishing Company; 2023.<br/><br/> 13. Komaroff A. Aripiprazole and lactation failure: the importance of shared decision making. A case report. Case Rep Womens Health. 2021;30:e00308. doi:10.1016/j.crwh.2021.e00308‌<br/><br/> 14. Dennis CL, McQueen K. Does maternal postpartum depressive symptomatology influence infant feeding outcomes? Acta Pediatr. 2007;96(4):590-594. doi:10.1111/j.1651-2227.2007.00184.x<br/><br/> 15. Chaput KH, Nettel-Aguirre A, Musto R, et al. Breastfeeding difficulties and supports and risk of postpartum depression in a cohort of women who have given birth in Calgary: a prospective cohort study. CMAJ Open. 2016;4(1):E103-E109. doi:10.9778/cmajo.20150009<br/><br/> 16. Dias CC, Figueiredo B. Breastfeeding and depression: a systematic review of the literature. J Affect Disord. 2015;171:142-154. doi:10.1016/j.jad.2014.09.022 <br/><br/> 17. Brown A, Rance J, Bennett P. Understanding the relationship between breastfeeding and postnatal depression: the role of pain and physical difficulties. J Adv Nurs. 2016;72(2):273-282. doi:10.1111/jan.12832<br/><br/> 18. Rosenbaum KA. Exclusive breastmilk pumping: a concept analysis. Nurs Forum. 2022;57(5):946-953. doi:10.1111/nuf.12766 <br/><br/> 19. Boone KM, Geraghty SR, Keim SA. Feeding at the breast and expressed milk feeding: associations with otitis media and diarrhea in infants. J Pediatr. 2016;174:118-125. doi:10.1016/j.jpeds.2016.04.006 <br/><br/> 20. Epstein LJ, Kristo D, Strollo PJ Jr, et al; Adult Obstructive Sleep Apnea Task Force of the American Academy of Sleep Medicine. Clinical guideline for the evaluation, management and long-term care of obstructive sleep apnea in adults. J Clin Sleep Med. 2009;5(3):263-276. <br/><br/> 21. Caan MP, Sreshta NE, Okwerekwu JA, et al. Clinical and legal considerations regarding breastfeeding on psychiatric units. J Am Acad Psychiatry Law. 2022;50(2):200-207. doi:10.29158/JAAPL.210086-21 <br/><br/> 22. Glangeaud-Freudenthal NMC, Rainelli C, Cazas O, et al. Inpatient mother and baby psychiatric units (MBUs) and day cares. In: Sutter-Dallay AL, Glangeaud-Freudenthal NC, Guedeney A, et al, eds. <i>Joint Care of Parents and Infants in Perinatal Psychiatry.</i> Springer, Cham; 2016:147-164. doi:10.1007/978-3-319-21557-0_10<br/><br/> 23. Dembosky A. A humane approach to caring for new mothers in psychiatric crisis. Health Aff (Millwood). 2021;40(10):1528-1533. doi:10.1377/hlthaff.2021.01288<br/><br/> 24. Connellan K, Bartholomaeus C, Due C, et al. A systematic review of research on psychiatric mother-baby units. Arch Womens Ment Health. 2017;20(3):373-388. doi:10.1007/s00737-017-0718-9<br/><br/> 25. Griffiths J, Lever Taylor B, Morant N, et al. A qualitative comparison of experiences of specialist mother and baby units versus general psychiatric wards. BMC Psychiatry. 2019;19(1):401. doi:10.1186/s12888-019-2389-8<br/><br/> 26. Heron J, Gilbert N, Dolman C, et al. Information and support needs during recovery from postpartum psychosis. Arch Womens Ment Health. 2012;15(3):155-165. doi:10.1007/s00737-012-0267-1 <br/><br/> 27. Robertson E, Lyons A. Living with puerperal psychosis: a qualitative analysis. Psychol Psychother. 2003;76(Pt 4):411-431. doi:10.1348/147608303770584755 <br/><br/> 28. Mental Welfare Commission for Scotland. <i>Perinatal Themed Visit Report: Keeping Mothers and Babies in Mind</i>. Mental Welfare Commission for Scotland; 2016.<br/><br/> 29. Wisner KL, Jennings KD, Conley B. Clinical dilemmas due to the lack of inpatient mother-baby units. Int J Psychiatry Med. 1996;26(4):479-493. doi:10.2190/NFJK-A4V7-CXUU-AM89<br/><br/> 30. Battle CL, Howard MM. A mother-baby psychiatric day hospital: history, rationale, and why perinatal mental health is important for obstetric medicine. Obstet Med. 2014;7(2):66-70. doi:10.1177/1753495X13514402<br/><br/> 31. Bullard ES, Meltzer-Brody S, Rubinow DR. The need for comprehensive psychiatric perinatal care-the University of North Carolina at Chapel Hill, Department of Psychiatry, Center for Women’s Mood Disorders launches the first dedicated inpatient program in the United States. Am J Obstet Gynecol. 2009;201(5):e10-e11. doi:10.1016/j.ajog.2009.05.004<br/><br/> 32. Meltzer-Brody S, Brandon AR, Pearson B, et al. Evaluating the clinical effectiveness of a specialized perinatal psychiatry inpatient unit. Arch Womens Ment Health. 2014;17(2):107-113. doi:10.1007/s00737-013-0390-7</p> <p class="reference"> 33. Alvarez-Toro V. Gender-specific care for women in psychiatric units. J Am Acad Psychiatry Law. 2022;JAAPL.220015-21. doi:10.29158/JAAPL.220015-21 <br/><br/> 34. Diaz-Caneja A, Johnson S. The views and experiences of severely mentally ill mothers--a qualitative study. Soc Psychiatry Psychiatr Epidemiol. 2004;39(6):472-482. doi:10.1007/s00127-004-0772-2<br/><br/> 35. Gewurtz R, Krupa T, Eastabrook S, et al. Prevalence and characteristics of parenting among people served by assertive community treatment. Psychiatr Rehabil J. 2004;28(1):63-65. doi:10.2975/28.2004.63.65 <br/><br/> 36. Howard LM, Kumar R, Thornicroft G. Psychosocial characteristics and needs of mothers with psychotic disorders. Br J Psychiatry. 2001;178:427-432. doi:10.1192/bjp.178.5.427 <br/><br/> 37. Hollingsworth LD. Child custody loss among women with persistent severe mental illness. Social Work Research. 2004;28(4):199-209. doi:10.1093/swr/28.4.199<br/><br/> 38. Dipple H, Smith S, Andrews H, et al. The experience of motherhood in women with severe and enduring mental illness. Soc Psychiatry Psychiatr Epidemiolf. 2002;37(7):336-340. doi:10.1007/s00127-002-0559-2<br/><br/> 39. Seeman MV. Intervention to prevent child custody loss in mothers with schizophrenia. Schizophr Res Treatment. 2012;2012:796763. doi:10.1155/2012/796763</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
1800267C.SIG
Disable zoom
Off

Opioid use disorder in pregnancy: A strategy for using methadone

Article Type
Changed
Tue, 12/12/2023 - 11:39
Display Headline
Opioid use disorder in pregnancy: A strategy for using methadone

In the United States, opioid use by patients who are pregnant more than quadrupled from 1999 to 2014.1 Opioid use disorder (OUD) in the perinatal period is associated with a higher risk for depression, suicide, malnutrition, domestic violence, and obstetric complications such as spontaneous abortion, preeclampsia, and premature delivery.2 Buprenorphine and methadone are the standard of care for treating OUD in pregnancy.3,4 While a literature review found that maternal treatment with buprenorphine has comparable efficacy to treatment with methadone,5 a small randomized, double-blind study found that compared to buprenorphine, methadone was associated with significantly lower use of additional opioids (P = .047).6 This suggests methadone has therapeutic value for patients who are pregnant.

Despite the benefits of methadone for treating perinatal OUD, the physiological changes that occur in patients who are pregnant—coupled with methadone’s unique pharmacologic properties—may complicate its use. Patients typically take methadone once a day, and the dose is titrated every 3 to 5 days to allow serum levels to reach steady state.7 During pregnancy, there are increases in both the volume of distribution and medication metabolism secondary to increased expression of the cytochrome P450 3A4 enzyme by the liver, intestine, and placenta.8 Additionally, as the pregnancy progresses, the rate of methadone metabolism increases.9 Methadone’s half-life (20 to 35 hours) leads to its accumulation in tissue and slow release into the blood.10 As a result, patients with OUD who are pregnant often require higher doses of methadone or divided dosing, particularly in the second and third trimesters.11

In this article, we provide a strategy for divided dosing of methadone for managing opioid withdrawal symptoms in the acute care setting. We present 2 cases of women with OUD who are pregnant and describe the collaboration of addiction medicine, consultation-liaison psychiatry, and obstetrics services.

CASE 1

Ms. H, age 29, is G3P2 and presents to the emergency department (ED) during her fourth pregnancy at 31 weeks, 1 day gestation. She has a history of opioid, cocaine, and benzodiazepine use disorders and chronic hepatitis C. Ms. H is enrolled in an opioid treatment program and takes methadone 190 mg/d in addition to nonprescribed opioids. In the ED, Ms. H requests medically supervised withdrawal management. Her urine toxicology is positive for cocaine, benzodiazepines, methadone, and opiates. Her laboratory results and electrocardiogram (ECG) are unremarkable. On admission, Ms. H’s Clinical Opiate Withdrawal Scale (COWS) score is 3, indicating minimal symptoms (5 to 12: mild; 13 to 24: moderate; 25 to 36: moderately severe; >36: severe). Fetal monitoring is reassuring.

Ms. H’s withdrawal is monitored with COWS every 4 hours. The treatment team initiates methadone 170 mg/d, with an additional 10 mg/d as needed to keep her COWS score <8, and daily QTc monitoring. Ms. H also receives lorazepam 2 to 4 mg/d as needed for benzodiazepine withdrawal. Despite the increase in her daily methadone dose, Ms. H continues to experience opioid withdrawal in the early evening and overnight. As a result, the treatment team increases Ms. H’s morning methadone dose to 190 mg and schedules an afternoon dose of 30 mg. Despite this adjustment, her COWS scores remain elevated in the afternoon and evening, and she requires additional as-needed doses of methadone. Methadone peak and trough levels are ordered to assess for rapid metabolism. The serum trough level is 190 ng/mL, which is low, and a serum peak level is not reported. Despite titration, Ms. H has a self-directed premature discharge.

Five days later at 32 weeks, 2 days gestation, Ms. H is readmitted after she had resumed use of opioids, benzodiazepines, and cocaine. Her vital signs are stable, and her laboratory results and ECG are unremarkable. Fetal monitoring is reassuring. Given Ms. H’s low methadone serum trough level and overall concern for rapid methadone metabolism, the treatment team decides to divide dosing of methadone. Over 9 days, the team titrates methadone to 170 mg twice daily on the day of discharge, which resolves Ms. H’s withdrawal symptoms.

At 38 weeks, 5 days gestation, Ms. H returns to the ED after experiencing labor contractions and opiate withdrawal symptoms after she resumed use of heroin, cocaine, and benzodiazepines. During this admission, Ms. H’s methadone is increased to 180 mg twice daily with additional as-needed doses for ongoing withdrawal symptoms. At 39 weeks, 2 days gestation, Ms. H has a scheduled cesarean delivery.

Her infant has a normal weight but is transferred to the neonatal intensive care unit (NICU) for management of neonatal opioid withdrawal syndrome (NOWS) and receives morphine. The baby remains in the NICU for 35 days and is discharged home without further treatment. When Ms. H is discharged, her methadone dose is 170 mg twice daily, which resolves her opioid withdrawal symptoms. The treatment team directs her to continue care in her methadone outpatient program and receive treatment for her cocaine and benzodiazepine use disorders. She declines residential or inpatient substance use treatment.

Continue to: CASE 2

 

 

CASE 2

Ms. M, age 39, is G4P2 and presents to the hospital during her fifth pregnancy at 27 weeks gestation. She has not received prenatal care for this pregnancy. She has a history of OUD and major depressive disorder (MDD). Ms. M’s urine toxicology is positive for opiates, fentanyl, and oxycodone. Her laboratory results are notable for mildly elevated alanine aminotransferase, positive hepatitis C antibody, and a hepatitis C viral load of 91,000, consistent with chronic hepatitis C infection. On admission, her COWS score is 14, indicating moderate withdrawal symptoms. Her ECG is unremarkable, and fetal monitoring is reassuring.

Ms. M had received methadone during a prior pregnancy and opts to reinitiate treatment with methadone during her current admission. The team initiates methadone 20 mg/d with additional as-needed doses for ongoing withdrawal symptoms. Due to a persistently elevated COWS score, Ms. M’s methadone is increased to 90 mg/d, which resolves her withdrawal symptoms. However, on Day 4, Ms. M reports having anxiety, refuses bloodwork to obtain methadone peak and trough levels, and prematurely discharges from the hospital.

One day later at 27 weeks, 5 days gestation, Ms. M is readmitted for continued management of opioid withdrawal. She presents with stable vital signs, an unremarkable ECG, and reassuring fetal monitoring. Her COWS score is 5. The treatment team reinitiates methadone at 80 mg/d and titrates it to 100 mg/d on Day 7. Given Ms. M’s ongoing evening cravings and concern for rapid methadone metabolism, on Day 10 the team switches the methadone dosing to 50 mg twice daily to maintain steady-state levels and promote patient comfort. Fluoxetine 20 mg/d is started for comorbid MDD and eventually increased to 80 mg/d. Ms. M is discharged on Day 15 with a regimen of methadone 60 mg/d in the morning and 70 mg/d at night. She plans to resume care in an opioid treatment program and follow up with psychiatry and hepatology for her anxiety and hepatitis C.

A need for aggressive treatment

Given the rising rates of opioid use by patients who are pregnant, harmful behavior related to opioid use, and a wealth of evidence supporting opioid agonist treatment for OUD in pregnancy, there is a growing need for guidance in managing perinatal OUD. A systematic approach to using methadone to treat OUD in patients who are pregnant is essential; the lack of data surrounding use of this medication in such patients may cause overall harm.12 Limited guidelines and a lack of familiarity with prescribing methadone to patients who are pregnant may lead clinicians to underdose patients, which can result in ongoing withdrawal, premature patient-directed discharges, and poor engagement in care.13 Both patients in the 2 cases described in this article experienced ongoing withdrawal symptoms despite daily titration of methadone. This suggests rapid metabolism, which was successfully managed by dividing the dosing of methadone, particularly in the latter trimesters.

These cases illustrate the need for aggressive perinatal opioid withdrawal management through rapid escalation of divided doses of methadone in a monitored acute care setting. Because methadone elimination is more rapid and clearance rates increase during the perinatal period, divided methadone dosing allows for sustained plasma methadone concentrations and improved outpatient treatment adherence.9,14,15

Continue to: Decreasing the rate of premature discharges

 

 

Decreasing the rate of premature discharges

In both cases, the patients discharged from the hospital prematurely, likely related to incomplete management of their opioid withdrawal or other withdrawal syndromes (both patients had multiple substance use disorders [SUDs]). Compared to patients without an SUD, patients with SUDs are 3 times more likely to have a self-directed discharge.16 Patients report leaving the hospital prematurely due to undertreated withdrawal, uncontrolled pain, discrimination by staff, and hospital restrictions.16 Recommendations to decrease the rates of premature patient-directed discharges in this population include providing patient-centered and harm reduction–oriented care in addition to adequate management of pain and withdrawal.17

Impact of methadone on fetal outcomes

Approximately 55% to 94% of infants born to patients who are opioid-dependent will develop NOWS. However, there is no relationship between this syndrome and therapeutic doses of methadone.18 Moreover, long-term research has found that after adjusting for socioeconomic factors, methadone treatment during pregnancy does not have an adverse effect on postnatal development. Divided dosing in maternal methadone administration is also shown to have less of an impact on fetal neurobehavior and NOWS.19

Our recommendations for methadone treatment for perinatal patients are outlined in the Table. Aggressive treatment of opioid withdrawal in the hospital can promote treatment engagement and prevent premature discharges. Clinicians should assess for other withdrawal syndromes when a patient has multiple SUDs and collaborate with an interdisciplinary team to improve patient outcomes.

CP02212e11_t1.png

Bottom Line

The prevalence of opioid use disorder (OUD) in patients who are pregnant is increasing. Methadone is an option for treating perinatal OUD, but the physiological changes that occur in patients who are pregnant—coupled with methadone’s unique pharmacologic properties—may complicate its use. Using divided doses of methadone can ensure the comfort and safety of the patient and their baby and improve adherence and outcomes.

Related Resources

Drug Brand Names

Buprenorphine • Buprenex, Suboxone, Zubsolv, Sublocade
Fentanyl • Abstral, Actiq
Fluoxetine • Prozac
Lorazepam • Ativan
Methadone • Methadose, Dolophine
Oxycodone • Oxycontin

[embed:render:related:node:259824]

References

1. Haight SC, Ko JY, Tong VT, et al. Opioid use disorder documented at delivery hospitalization – United States, 1999-2014. MMWR Morb Mortal Wkly Rep. 2018;67(31):845-849.

2. Kaltenbach K, Berghella V, Finnegan L. Opioid dependence during pregnancy. Effects and management. Obstet Gynecol Clin North Am. 1998;25(1):139-151. doi:10.1016/S0889-8545(05)70362-4

3. Baumgaertner E. Biden administration offers plan to get addiction-fighting medicine to pregnant women. The New York Times. October 21, 2022. Accessed February 23, 2023. https://www.nytimes.com/2022/10/21/health/addiction-treatment-pregnancy.html

4. Jones HE, Fischer G, Heil SH, et al. Maternal Opioid Treatment: Human Experimental Research (MOTHER)--approach, issues and lessons learned. Addiction. 2012;107 Suppl 1(0 1):28-35. doi:10.1111/j.1360-0443.2012.04036.x

5. Jones HE, Heil SH, Baewert A, et al. Buprenorphine treatment of opioid-dependent pregnant women: a comprehensive review. Addiction. 2012;107 Suppl 1:5-27.

6. Fischer G, Ortner R, Rohrmeister K, et al. Methadone versus buprenorphine in pregnant addicts: a double-blind, double-dummy comparison study. Addiction. 2006;101(2):275-281. doi:10.1111/j.1360-0443.2006.01321.x

7. Substance Abuse and Mental Health Services Administration. Chapter 3B: Methadone. Medications for Opioid Use Disorder: For Healthcare and Addiction Professionals, Policymakers, Patients, and Families: Updated 2021. Substance Abuse and Mental Health Services Administration; August 2021. https://www.ncbi.nlm.nih.gov/books/NBK574918/

8. Feghali M, Venkataramanan R, Caritis S. Pharmacokinetics of drugs in pregnancy. Semin Perinatol. 2015;39(7):512-519. doi:10.1053/j.semperi.2015.08.003

9. McCarthy JJ, Vasti EJ, Leamon MH, et al. The use of serum methadone/metabolite ratios to monitor changing perinatal pharmacokinetics. J Addict Med. 2018;12(3): 241-246.

10. Center for Substance Abuse Treatment. Medication-Assisted Treatment for Opioid Addiction in Opioid Treatment Programs. Treatment Improvement Protocol Series No. 43. Substance Abuse and Mental Health Service Administration; 2005.

11. Substance Abuse and Mental Health Services Administration. Clinical Guidance for Treating Pregnant and Parenting Women with Opioid Use Disorder and Their Infants. Createspace Independent Publishing Platform; 2018.

12. Balch B. Prescribing without data: doctors advocate for the inclusion of pregnant people in clinical research. Association of American Medical Colleges. March 22, 2022. Accessed September 30, 2022. https://www.aamc.org/news-insights/prescribing-without-data-doctors-advocate-inclusion-pregnant-people-clinical-research

13. Leavitt SB. Methadone Dosing & Safety in the Treatment of Opioid Addiction. 2003. Addiction Treatment Forum. Accessed November 28, 2023. https://atforum.com/documents/DosingandSafetyWP.pdf

14. McCarthy JJ, Leamon MH, Willitts NH, et al. The effect of methadone dose regimen on neonatal abstinence syndrome. J Addict Med. 2015; 9(2):105-110.

15. DePetrillo PB, Rice JM. Methadone dosing and pregnancy: impact on program compliance. Int J Addict. 1995;30(2):207-217.

16. Simon R, Snow R, Wakeman S. Understanding why patients with substance use disorders leave the hospital against medical advice: a qualitative study. Subst Abus. 2020;41(4):519-525. doi:10.1080/08897077.2019.1671942

17. McNeil R, Small W, Wood E, et al. Hospitals as a ‘risk environment’: an ethno-epidemiological study of voluntary and involuntary discharge from hospital against medical advice among people who inject drugs. Soc Sci Med. 2014;105:59-66.

18. Jones HE, Jansson LM, O’Grady KE, et al. The relationship between maternal methadone dose at delivery and neonatal outcome: methodological and design considerations. Neurotoxicol Teratol. 2013;39:110-115.

19. McCarthy JJ, Leamon MH, Parr MS, et al. High-dose methadone maintenance in pregnancy: maternal and neonatal outcomes. Am J Obstet Gynecol. 2005;193(3 Pt 1):606-610.

Article PDF
Author and Disclosure Information

Sitara Soundararajan, MD
PGY-3 Psychiatry Resident
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center
Bronx, New York

Kristine Torres-Lockhart, MD
Assistant Professor
Department of Medicine and Department of Psychiatry and Behavioral Sciences
Albert Einstein College of Medicine
Bronx, New York

Rubiahna Vaughn, MD, MPH
Assistant Professor
Department of Psychiatry and Behavioral Services
Albert Einstein College of Medicine
Director, Consultation-Liaison and Emergency Psychiatry
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center, Weiler Hospital
Bronx, New York

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Issue
Current Psychiatry - 22(12)
Publications
Topics
Page Number
e11-e15
Sections
Author and Disclosure Information

Sitara Soundararajan, MD
PGY-3 Psychiatry Resident
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center
Bronx, New York

Kristine Torres-Lockhart, MD
Assistant Professor
Department of Medicine and Department of Psychiatry and Behavioral Sciences
Albert Einstein College of Medicine
Bronx, New York

Rubiahna Vaughn, MD, MPH
Assistant Professor
Department of Psychiatry and Behavioral Services
Albert Einstein College of Medicine
Director, Consultation-Liaison and Emergency Psychiatry
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center, Weiler Hospital
Bronx, New York

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Author and Disclosure Information

Sitara Soundararajan, MD
PGY-3 Psychiatry Resident
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center
Bronx, New York

Kristine Torres-Lockhart, MD
Assistant Professor
Department of Medicine and Department of Psychiatry and Behavioral Sciences
Albert Einstein College of Medicine
Bronx, New York

Rubiahna Vaughn, MD, MPH
Assistant Professor
Department of Psychiatry and Behavioral Services
Albert Einstein College of Medicine
Director, Consultation-Liaison and Emergency Psychiatry
Department of Psychiatry and Behavioral Sciences
Montefiore Medical Center, Weiler Hospital
Bronx, New York

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Article PDF
Article PDF

In the United States, opioid use by patients who are pregnant more than quadrupled from 1999 to 2014.1 Opioid use disorder (OUD) in the perinatal period is associated with a higher risk for depression, suicide, malnutrition, domestic violence, and obstetric complications such as spontaneous abortion, preeclampsia, and premature delivery.2 Buprenorphine and methadone are the standard of care for treating OUD in pregnancy.3,4 While a literature review found that maternal treatment with buprenorphine has comparable efficacy to treatment with methadone,5 a small randomized, double-blind study found that compared to buprenorphine, methadone was associated with significantly lower use of additional opioids (P = .047).6 This suggests methadone has therapeutic value for patients who are pregnant.

Despite the benefits of methadone for treating perinatal OUD, the physiological changes that occur in patients who are pregnant—coupled with methadone’s unique pharmacologic properties—may complicate its use. Patients typically take methadone once a day, and the dose is titrated every 3 to 5 days to allow serum levels to reach steady state.7 During pregnancy, there are increases in both the volume of distribution and medication metabolism secondary to increased expression of the cytochrome P450 3A4 enzyme by the liver, intestine, and placenta.8 Additionally, as the pregnancy progresses, the rate of methadone metabolism increases.9 Methadone’s half-life (20 to 35 hours) leads to its accumulation in tissue and slow release into the blood.10 As a result, patients with OUD who are pregnant often require higher doses of methadone or divided dosing, particularly in the second and third trimesters.11

In this article, we provide a strategy for divided dosing of methadone for managing opioid withdrawal symptoms in the acute care setting. We present 2 cases of women with OUD who are pregnant and describe the collaboration of addiction medicine, consultation-liaison psychiatry, and obstetrics services.

CASE 1

Ms. H, age 29, is G3P2 and presents to the emergency department (ED) during her fourth pregnancy at 31 weeks, 1 day gestation. She has a history of opioid, cocaine, and benzodiazepine use disorders and chronic hepatitis C. Ms. H is enrolled in an opioid treatment program and takes methadone 190 mg/d in addition to nonprescribed opioids. In the ED, Ms. H requests medically supervised withdrawal management. Her urine toxicology is positive for cocaine, benzodiazepines, methadone, and opiates. Her laboratory results and electrocardiogram (ECG) are unremarkable. On admission, Ms. H’s Clinical Opiate Withdrawal Scale (COWS) score is 3, indicating minimal symptoms (5 to 12: mild; 13 to 24: moderate; 25 to 36: moderately severe; >36: severe). Fetal monitoring is reassuring.

Ms. H’s withdrawal is monitored with COWS every 4 hours. The treatment team initiates methadone 170 mg/d, with an additional 10 mg/d as needed to keep her COWS score <8, and daily QTc monitoring. Ms. H also receives lorazepam 2 to 4 mg/d as needed for benzodiazepine withdrawal. Despite the increase in her daily methadone dose, Ms. H continues to experience opioid withdrawal in the early evening and overnight. As a result, the treatment team increases Ms. H’s morning methadone dose to 190 mg and schedules an afternoon dose of 30 mg. Despite this adjustment, her COWS scores remain elevated in the afternoon and evening, and she requires additional as-needed doses of methadone. Methadone peak and trough levels are ordered to assess for rapid metabolism. The serum trough level is 190 ng/mL, which is low, and a serum peak level is not reported. Despite titration, Ms. H has a self-directed premature discharge.

Five days later at 32 weeks, 2 days gestation, Ms. H is readmitted after she had resumed use of opioids, benzodiazepines, and cocaine. Her vital signs are stable, and her laboratory results and ECG are unremarkable. Fetal monitoring is reassuring. Given Ms. H’s low methadone serum trough level and overall concern for rapid methadone metabolism, the treatment team decides to divide dosing of methadone. Over 9 days, the team titrates methadone to 170 mg twice daily on the day of discharge, which resolves Ms. H’s withdrawal symptoms.

At 38 weeks, 5 days gestation, Ms. H returns to the ED after experiencing labor contractions and opiate withdrawal symptoms after she resumed use of heroin, cocaine, and benzodiazepines. During this admission, Ms. H’s methadone is increased to 180 mg twice daily with additional as-needed doses for ongoing withdrawal symptoms. At 39 weeks, 2 days gestation, Ms. H has a scheduled cesarean delivery.

Her infant has a normal weight but is transferred to the neonatal intensive care unit (NICU) for management of neonatal opioid withdrawal syndrome (NOWS) and receives morphine. The baby remains in the NICU for 35 days and is discharged home without further treatment. When Ms. H is discharged, her methadone dose is 170 mg twice daily, which resolves her opioid withdrawal symptoms. The treatment team directs her to continue care in her methadone outpatient program and receive treatment for her cocaine and benzodiazepine use disorders. She declines residential or inpatient substance use treatment.

Continue to: CASE 2

 

 

CASE 2

Ms. M, age 39, is G4P2 and presents to the hospital during her fifth pregnancy at 27 weeks gestation. She has not received prenatal care for this pregnancy. She has a history of OUD and major depressive disorder (MDD). Ms. M’s urine toxicology is positive for opiates, fentanyl, and oxycodone. Her laboratory results are notable for mildly elevated alanine aminotransferase, positive hepatitis C antibody, and a hepatitis C viral load of 91,000, consistent with chronic hepatitis C infection. On admission, her COWS score is 14, indicating moderate withdrawal symptoms. Her ECG is unremarkable, and fetal monitoring is reassuring.

Ms. M had received methadone during a prior pregnancy and opts to reinitiate treatment with methadone during her current admission. The team initiates methadone 20 mg/d with additional as-needed doses for ongoing withdrawal symptoms. Due to a persistently elevated COWS score, Ms. M’s methadone is increased to 90 mg/d, which resolves her withdrawal symptoms. However, on Day 4, Ms. M reports having anxiety, refuses bloodwork to obtain methadone peak and trough levels, and prematurely discharges from the hospital.

One day later at 27 weeks, 5 days gestation, Ms. M is readmitted for continued management of opioid withdrawal. She presents with stable vital signs, an unremarkable ECG, and reassuring fetal monitoring. Her COWS score is 5. The treatment team reinitiates methadone at 80 mg/d and titrates it to 100 mg/d on Day 7. Given Ms. M’s ongoing evening cravings and concern for rapid methadone metabolism, on Day 10 the team switches the methadone dosing to 50 mg twice daily to maintain steady-state levels and promote patient comfort. Fluoxetine 20 mg/d is started for comorbid MDD and eventually increased to 80 mg/d. Ms. M is discharged on Day 15 with a regimen of methadone 60 mg/d in the morning and 70 mg/d at night. She plans to resume care in an opioid treatment program and follow up with psychiatry and hepatology for her anxiety and hepatitis C.

A need for aggressive treatment

Given the rising rates of opioid use by patients who are pregnant, harmful behavior related to opioid use, and a wealth of evidence supporting opioid agonist treatment for OUD in pregnancy, there is a growing need for guidance in managing perinatal OUD. A systematic approach to using methadone to treat OUD in patients who are pregnant is essential; the lack of data surrounding use of this medication in such patients may cause overall harm.12 Limited guidelines and a lack of familiarity with prescribing methadone to patients who are pregnant may lead clinicians to underdose patients, which can result in ongoing withdrawal, premature patient-directed discharges, and poor engagement in care.13 Both patients in the 2 cases described in this article experienced ongoing withdrawal symptoms despite daily titration of methadone. This suggests rapid metabolism, which was successfully managed by dividing the dosing of methadone, particularly in the latter trimesters.

These cases illustrate the need for aggressive perinatal opioid withdrawal management through rapid escalation of divided doses of methadone in a monitored acute care setting. Because methadone elimination is more rapid and clearance rates increase during the perinatal period, divided methadone dosing allows for sustained plasma methadone concentrations and improved outpatient treatment adherence.9,14,15

Continue to: Decreasing the rate of premature discharges

 

 

Decreasing the rate of premature discharges

In both cases, the patients discharged from the hospital prematurely, likely related to incomplete management of their opioid withdrawal or other withdrawal syndromes (both patients had multiple substance use disorders [SUDs]). Compared to patients without an SUD, patients with SUDs are 3 times more likely to have a self-directed discharge.16 Patients report leaving the hospital prematurely due to undertreated withdrawal, uncontrolled pain, discrimination by staff, and hospital restrictions.16 Recommendations to decrease the rates of premature patient-directed discharges in this population include providing patient-centered and harm reduction–oriented care in addition to adequate management of pain and withdrawal.17

Impact of methadone on fetal outcomes

Approximately 55% to 94% of infants born to patients who are opioid-dependent will develop NOWS. However, there is no relationship between this syndrome and therapeutic doses of methadone.18 Moreover, long-term research has found that after adjusting for socioeconomic factors, methadone treatment during pregnancy does not have an adverse effect on postnatal development. Divided dosing in maternal methadone administration is also shown to have less of an impact on fetal neurobehavior and NOWS.19

Our recommendations for methadone treatment for perinatal patients are outlined in the Table. Aggressive treatment of opioid withdrawal in the hospital can promote treatment engagement and prevent premature discharges. Clinicians should assess for other withdrawal syndromes when a patient has multiple SUDs and collaborate with an interdisciplinary team to improve patient outcomes.

CP02212e11_t1.png

Bottom Line

The prevalence of opioid use disorder (OUD) in patients who are pregnant is increasing. Methadone is an option for treating perinatal OUD, but the physiological changes that occur in patients who are pregnant—coupled with methadone’s unique pharmacologic properties—may complicate its use. Using divided doses of methadone can ensure the comfort and safety of the patient and their baby and improve adherence and outcomes.

Related Resources

Drug Brand Names

Buprenorphine • Buprenex, Suboxone, Zubsolv, Sublocade
Fentanyl • Abstral, Actiq
Fluoxetine • Prozac
Lorazepam • Ativan
Methadone • Methadose, Dolophine
Oxycodone • Oxycontin

[embed:render:related:node:259824]

In the United States, opioid use by patients who are pregnant more than quadrupled from 1999 to 2014.1 Opioid use disorder (OUD) in the perinatal period is associated with a higher risk for depression, suicide, malnutrition, domestic violence, and obstetric complications such as spontaneous abortion, preeclampsia, and premature delivery.2 Buprenorphine and methadone are the standard of care for treating OUD in pregnancy.3,4 While a literature review found that maternal treatment with buprenorphine has comparable efficacy to treatment with methadone,5 a small randomized, double-blind study found that compared to buprenorphine, methadone was associated with significantly lower use of additional opioids (P = .047).6 This suggests methadone has therapeutic value for patients who are pregnant.

Despite the benefits of methadone for treating perinatal OUD, the physiological changes that occur in patients who are pregnant—coupled with methadone’s unique pharmacologic properties—may complicate its use. Patients typically take methadone once a day, and the dose is titrated every 3 to 5 days to allow serum levels to reach steady state.7 During pregnancy, there are increases in both the volume of distribution and medication metabolism secondary to increased expression of the cytochrome P450 3A4 enzyme by the liver, intestine, and placenta.8 Additionally, as the pregnancy progresses, the rate of methadone metabolism increases.9 Methadone’s half-life (20 to 35 hours) leads to its accumulation in tissue and slow release into the blood.10 As a result, patients with OUD who are pregnant often require higher doses of methadone or divided dosing, particularly in the second and third trimesters.11

In this article, we provide a strategy for divided dosing of methadone for managing opioid withdrawal symptoms in the acute care setting. We present 2 cases of women with OUD who are pregnant and describe the collaboration of addiction medicine, consultation-liaison psychiatry, and obstetrics services.

CASE 1

Ms. H, age 29, is G3P2 and presents to the emergency department (ED) during her fourth pregnancy at 31 weeks, 1 day gestation. She has a history of opioid, cocaine, and benzodiazepine use disorders and chronic hepatitis C. Ms. H is enrolled in an opioid treatment program and takes methadone 190 mg/d in addition to nonprescribed opioids. In the ED, Ms. H requests medically supervised withdrawal management. Her urine toxicology is positive for cocaine, benzodiazepines, methadone, and opiates. Her laboratory results and electrocardiogram (ECG) are unremarkable. On admission, Ms. H’s Clinical Opiate Withdrawal Scale (COWS) score is 3, indicating minimal symptoms (5 to 12: mild; 13 to 24: moderate; 25 to 36: moderately severe; >36: severe). Fetal monitoring is reassuring.

Ms. H’s withdrawal is monitored with COWS every 4 hours. The treatment team initiates methadone 170 mg/d, with an additional 10 mg/d as needed to keep her COWS score <8, and daily QTc monitoring. Ms. H also receives lorazepam 2 to 4 mg/d as needed for benzodiazepine withdrawal. Despite the increase in her daily methadone dose, Ms. H continues to experience opioid withdrawal in the early evening and overnight. As a result, the treatment team increases Ms. H’s morning methadone dose to 190 mg and schedules an afternoon dose of 30 mg. Despite this adjustment, her COWS scores remain elevated in the afternoon and evening, and she requires additional as-needed doses of methadone. Methadone peak and trough levels are ordered to assess for rapid metabolism. The serum trough level is 190 ng/mL, which is low, and a serum peak level is not reported. Despite titration, Ms. H has a self-directed premature discharge.

Five days later at 32 weeks, 2 days gestation, Ms. H is readmitted after she had resumed use of opioids, benzodiazepines, and cocaine. Her vital signs are stable, and her laboratory results and ECG are unremarkable. Fetal monitoring is reassuring. Given Ms. H’s low methadone serum trough level and overall concern for rapid methadone metabolism, the treatment team decides to divide dosing of methadone. Over 9 days, the team titrates methadone to 170 mg twice daily on the day of discharge, which resolves Ms. H’s withdrawal symptoms.

At 38 weeks, 5 days gestation, Ms. H returns to the ED after experiencing labor contractions and opiate withdrawal symptoms after she resumed use of heroin, cocaine, and benzodiazepines. During this admission, Ms. H’s methadone is increased to 180 mg twice daily with additional as-needed doses for ongoing withdrawal symptoms. At 39 weeks, 2 days gestation, Ms. H has a scheduled cesarean delivery.

Her infant has a normal weight but is transferred to the neonatal intensive care unit (NICU) for management of neonatal opioid withdrawal syndrome (NOWS) and receives morphine. The baby remains in the NICU for 35 days and is discharged home without further treatment. When Ms. H is discharged, her methadone dose is 170 mg twice daily, which resolves her opioid withdrawal symptoms. The treatment team directs her to continue care in her methadone outpatient program and receive treatment for her cocaine and benzodiazepine use disorders. She declines residential or inpatient substance use treatment.

Continue to: CASE 2

 

 

CASE 2

Ms. M, age 39, is G4P2 and presents to the hospital during her fifth pregnancy at 27 weeks gestation. She has not received prenatal care for this pregnancy. She has a history of OUD and major depressive disorder (MDD). Ms. M’s urine toxicology is positive for opiates, fentanyl, and oxycodone. Her laboratory results are notable for mildly elevated alanine aminotransferase, positive hepatitis C antibody, and a hepatitis C viral load of 91,000, consistent with chronic hepatitis C infection. On admission, her COWS score is 14, indicating moderate withdrawal symptoms. Her ECG is unremarkable, and fetal monitoring is reassuring.

Ms. M had received methadone during a prior pregnancy and opts to reinitiate treatment with methadone during her current admission. The team initiates methadone 20 mg/d with additional as-needed doses for ongoing withdrawal symptoms. Due to a persistently elevated COWS score, Ms. M’s methadone is increased to 90 mg/d, which resolves her withdrawal symptoms. However, on Day 4, Ms. M reports having anxiety, refuses bloodwork to obtain methadone peak and trough levels, and prematurely discharges from the hospital.

One day later at 27 weeks, 5 days gestation, Ms. M is readmitted for continued management of opioid withdrawal. She presents with stable vital signs, an unremarkable ECG, and reassuring fetal monitoring. Her COWS score is 5. The treatment team reinitiates methadone at 80 mg/d and titrates it to 100 mg/d on Day 7. Given Ms. M’s ongoing evening cravings and concern for rapid methadone metabolism, on Day 10 the team switches the methadone dosing to 50 mg twice daily to maintain steady-state levels and promote patient comfort. Fluoxetine 20 mg/d is started for comorbid MDD and eventually increased to 80 mg/d. Ms. M is discharged on Day 15 with a regimen of methadone 60 mg/d in the morning and 70 mg/d at night. She plans to resume care in an opioid treatment program and follow up with psychiatry and hepatology for her anxiety and hepatitis C.

A need for aggressive treatment

Given the rising rates of opioid use by patients who are pregnant, harmful behavior related to opioid use, and a wealth of evidence supporting opioid agonist treatment for OUD in pregnancy, there is a growing need for guidance in managing perinatal OUD. A systematic approach to using methadone to treat OUD in patients who are pregnant is essential; the lack of data surrounding use of this medication in such patients may cause overall harm.12 Limited guidelines and a lack of familiarity with prescribing methadone to patients who are pregnant may lead clinicians to underdose patients, which can result in ongoing withdrawal, premature patient-directed discharges, and poor engagement in care.13 Both patients in the 2 cases described in this article experienced ongoing withdrawal symptoms despite daily titration of methadone. This suggests rapid metabolism, which was successfully managed by dividing the dosing of methadone, particularly in the latter trimesters.

These cases illustrate the need for aggressive perinatal opioid withdrawal management through rapid escalation of divided doses of methadone in a monitored acute care setting. Because methadone elimination is more rapid and clearance rates increase during the perinatal period, divided methadone dosing allows for sustained plasma methadone concentrations and improved outpatient treatment adherence.9,14,15

Continue to: Decreasing the rate of premature discharges

 

 

Decreasing the rate of premature discharges

In both cases, the patients discharged from the hospital prematurely, likely related to incomplete management of their opioid withdrawal or other withdrawal syndromes (both patients had multiple substance use disorders [SUDs]). Compared to patients without an SUD, patients with SUDs are 3 times more likely to have a self-directed discharge.16 Patients report leaving the hospital prematurely due to undertreated withdrawal, uncontrolled pain, discrimination by staff, and hospital restrictions.16 Recommendations to decrease the rates of premature patient-directed discharges in this population include providing patient-centered and harm reduction–oriented care in addition to adequate management of pain and withdrawal.17

Impact of methadone on fetal outcomes

Approximately 55% to 94% of infants born to patients who are opioid-dependent will develop NOWS. However, there is no relationship between this syndrome and therapeutic doses of methadone.18 Moreover, long-term research has found that after adjusting for socioeconomic factors, methadone treatment during pregnancy does not have an adverse effect on postnatal development. Divided dosing in maternal methadone administration is also shown to have less of an impact on fetal neurobehavior and NOWS.19

Our recommendations for methadone treatment for perinatal patients are outlined in the Table. Aggressive treatment of opioid withdrawal in the hospital can promote treatment engagement and prevent premature discharges. Clinicians should assess for other withdrawal syndromes when a patient has multiple SUDs and collaborate with an interdisciplinary team to improve patient outcomes.

CP02212e11_t1.png

Bottom Line

The prevalence of opioid use disorder (OUD) in patients who are pregnant is increasing. Methadone is an option for treating perinatal OUD, but the physiological changes that occur in patients who are pregnant—coupled with methadone’s unique pharmacologic properties—may complicate its use. Using divided doses of methadone can ensure the comfort and safety of the patient and their baby and improve adherence and outcomes.

Related Resources

Drug Brand Names

Buprenorphine • Buprenex, Suboxone, Zubsolv, Sublocade
Fentanyl • Abstral, Actiq
Fluoxetine • Prozac
Lorazepam • Ativan
Methadone • Methadose, Dolophine
Oxycodone • Oxycontin

[embed:render:related:node:259824]

References

1. Haight SC, Ko JY, Tong VT, et al. Opioid use disorder documented at delivery hospitalization – United States, 1999-2014. MMWR Morb Mortal Wkly Rep. 2018;67(31):845-849.

2. Kaltenbach K, Berghella V, Finnegan L. Opioid dependence during pregnancy. Effects and management. Obstet Gynecol Clin North Am. 1998;25(1):139-151. doi:10.1016/S0889-8545(05)70362-4

3. Baumgaertner E. Biden administration offers plan to get addiction-fighting medicine to pregnant women. The New York Times. October 21, 2022. Accessed February 23, 2023. https://www.nytimes.com/2022/10/21/health/addiction-treatment-pregnancy.html

4. Jones HE, Fischer G, Heil SH, et al. Maternal Opioid Treatment: Human Experimental Research (MOTHER)--approach, issues and lessons learned. Addiction. 2012;107 Suppl 1(0 1):28-35. doi:10.1111/j.1360-0443.2012.04036.x

5. Jones HE, Heil SH, Baewert A, et al. Buprenorphine treatment of opioid-dependent pregnant women: a comprehensive review. Addiction. 2012;107 Suppl 1:5-27.

6. Fischer G, Ortner R, Rohrmeister K, et al. Methadone versus buprenorphine in pregnant addicts: a double-blind, double-dummy comparison study. Addiction. 2006;101(2):275-281. doi:10.1111/j.1360-0443.2006.01321.x

7. Substance Abuse and Mental Health Services Administration. Chapter 3B: Methadone. Medications for Opioid Use Disorder: For Healthcare and Addiction Professionals, Policymakers, Patients, and Families: Updated 2021. Substance Abuse and Mental Health Services Administration; August 2021. https://www.ncbi.nlm.nih.gov/books/NBK574918/

8. Feghali M, Venkataramanan R, Caritis S. Pharmacokinetics of drugs in pregnancy. Semin Perinatol. 2015;39(7):512-519. doi:10.1053/j.semperi.2015.08.003

9. McCarthy JJ, Vasti EJ, Leamon MH, et al. The use of serum methadone/metabolite ratios to monitor changing perinatal pharmacokinetics. J Addict Med. 2018;12(3): 241-246.

10. Center for Substance Abuse Treatment. Medication-Assisted Treatment for Opioid Addiction in Opioid Treatment Programs. Treatment Improvement Protocol Series No. 43. Substance Abuse and Mental Health Service Administration; 2005.

11. Substance Abuse and Mental Health Services Administration. Clinical Guidance for Treating Pregnant and Parenting Women with Opioid Use Disorder and Their Infants. Createspace Independent Publishing Platform; 2018.

12. Balch B. Prescribing without data: doctors advocate for the inclusion of pregnant people in clinical research. Association of American Medical Colleges. March 22, 2022. Accessed September 30, 2022. https://www.aamc.org/news-insights/prescribing-without-data-doctors-advocate-inclusion-pregnant-people-clinical-research

13. Leavitt SB. Methadone Dosing & Safety in the Treatment of Opioid Addiction. 2003. Addiction Treatment Forum. Accessed November 28, 2023. https://atforum.com/documents/DosingandSafetyWP.pdf

14. McCarthy JJ, Leamon MH, Willitts NH, et al. The effect of methadone dose regimen on neonatal abstinence syndrome. J Addict Med. 2015; 9(2):105-110.

15. DePetrillo PB, Rice JM. Methadone dosing and pregnancy: impact on program compliance. Int J Addict. 1995;30(2):207-217.

16. Simon R, Snow R, Wakeman S. Understanding why patients with substance use disorders leave the hospital against medical advice: a qualitative study. Subst Abus. 2020;41(4):519-525. doi:10.1080/08897077.2019.1671942

17. McNeil R, Small W, Wood E, et al. Hospitals as a ‘risk environment’: an ethno-epidemiological study of voluntary and involuntary discharge from hospital against medical advice among people who inject drugs. Soc Sci Med. 2014;105:59-66.

18. Jones HE, Jansson LM, O’Grady KE, et al. The relationship between maternal methadone dose at delivery and neonatal outcome: methodological and design considerations. Neurotoxicol Teratol. 2013;39:110-115.

19. McCarthy JJ, Leamon MH, Parr MS, et al. High-dose methadone maintenance in pregnancy: maternal and neonatal outcomes. Am J Obstet Gynecol. 2005;193(3 Pt 1):606-610.

References

1. Haight SC, Ko JY, Tong VT, et al. Opioid use disorder documented at delivery hospitalization – United States, 1999-2014. MMWR Morb Mortal Wkly Rep. 2018;67(31):845-849.

2. Kaltenbach K, Berghella V, Finnegan L. Opioid dependence during pregnancy. Effects and management. Obstet Gynecol Clin North Am. 1998;25(1):139-151. doi:10.1016/S0889-8545(05)70362-4

3. Baumgaertner E. Biden administration offers plan to get addiction-fighting medicine to pregnant women. The New York Times. October 21, 2022. Accessed February 23, 2023. https://www.nytimes.com/2022/10/21/health/addiction-treatment-pregnancy.html

4. Jones HE, Fischer G, Heil SH, et al. Maternal Opioid Treatment: Human Experimental Research (MOTHER)--approach, issues and lessons learned. Addiction. 2012;107 Suppl 1(0 1):28-35. doi:10.1111/j.1360-0443.2012.04036.x

5. Jones HE, Heil SH, Baewert A, et al. Buprenorphine treatment of opioid-dependent pregnant women: a comprehensive review. Addiction. 2012;107 Suppl 1:5-27.

6. Fischer G, Ortner R, Rohrmeister K, et al. Methadone versus buprenorphine in pregnant addicts: a double-blind, double-dummy comparison study. Addiction. 2006;101(2):275-281. doi:10.1111/j.1360-0443.2006.01321.x

7. Substance Abuse and Mental Health Services Administration. Chapter 3B: Methadone. Medications for Opioid Use Disorder: For Healthcare and Addiction Professionals, Policymakers, Patients, and Families: Updated 2021. Substance Abuse and Mental Health Services Administration; August 2021. https://www.ncbi.nlm.nih.gov/books/NBK574918/

8. Feghali M, Venkataramanan R, Caritis S. Pharmacokinetics of drugs in pregnancy. Semin Perinatol. 2015;39(7):512-519. doi:10.1053/j.semperi.2015.08.003

9. McCarthy JJ, Vasti EJ, Leamon MH, et al. The use of serum methadone/metabolite ratios to monitor changing perinatal pharmacokinetics. J Addict Med. 2018;12(3): 241-246.

10. Center for Substance Abuse Treatment. Medication-Assisted Treatment for Opioid Addiction in Opioid Treatment Programs. Treatment Improvement Protocol Series No. 43. Substance Abuse and Mental Health Service Administration; 2005.

11. Substance Abuse and Mental Health Services Administration. Clinical Guidance for Treating Pregnant and Parenting Women with Opioid Use Disorder and Their Infants. Createspace Independent Publishing Platform; 2018.

12. Balch B. Prescribing without data: doctors advocate for the inclusion of pregnant people in clinical research. Association of American Medical Colleges. March 22, 2022. Accessed September 30, 2022. https://www.aamc.org/news-insights/prescribing-without-data-doctors-advocate-inclusion-pregnant-people-clinical-research

13. Leavitt SB. Methadone Dosing & Safety in the Treatment of Opioid Addiction. 2003. Addiction Treatment Forum. Accessed November 28, 2023. https://atforum.com/documents/DosingandSafetyWP.pdf

14. McCarthy JJ, Leamon MH, Willitts NH, et al. The effect of methadone dose regimen on neonatal abstinence syndrome. J Addict Med. 2015; 9(2):105-110.

15. DePetrillo PB, Rice JM. Methadone dosing and pregnancy: impact on program compliance. Int J Addict. 1995;30(2):207-217.

16. Simon R, Snow R, Wakeman S. Understanding why patients with substance use disorders leave the hospital against medical advice: a qualitative study. Subst Abus. 2020;41(4):519-525. doi:10.1080/08897077.2019.1671942

17. McNeil R, Small W, Wood E, et al. Hospitals as a ‘risk environment’: an ethno-epidemiological study of voluntary and involuntary discharge from hospital against medical advice among people who inject drugs. Soc Sci Med. 2014;105:59-66.

18. Jones HE, Jansson LM, O’Grady KE, et al. The relationship between maternal methadone dose at delivery and neonatal outcome: methodological and design considerations. Neurotoxicol Teratol. 2013;39:110-115.

19. McCarthy JJ, Leamon MH, Parr MS, et al. High-dose methadone maintenance in pregnancy: maternal and neonatal outcomes. Am J Obstet Gynecol. 2005;193(3 Pt 1):606-610.

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
e11-e15
Page Number
e11-e15
Publications
Publications
Topics
Article Type
Display Headline
Opioid use disorder in pregnancy: A strategy for using methadone
Display Headline
Opioid use disorder in pregnancy: A strategy for using methadone
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1223 Soundararajan OUD</fileName> <TBEID>0C02ED63.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02ED63</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Opioid use disorder in pregnancy</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231212T065034</firstPublished> <LastPublished>20231212T065034</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231212T065033</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Sitara Soundararajan, MD, Kristine Torres-Lockhart, MD</byline> <bylineText/> <bylineFull>Sitara Soundararajan, MD, Kristine Torres-Lockhart, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>e11-e15</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>In the United States, opioid use by patients who are pregnant more than quadrupled from 1999 to 2014.1 Opioid use disorder (OUD) in the perinatal period is asso</metaDescription> <articlePDF>299601</articlePDF> <teaserImage/> <title>Opioid use disorder in pregnancy: A strategy for using methadone</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords> <keyword>opioid use disorder</keyword> <keyword> OUD</keyword> <keyword> pregnancy</keyword> <keyword> addiction medicine</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">26935</term> </sections> <topics> <term canonical="true">174</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/1800267d.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Opioid use disorder in pregnancy: A strategy for using methadone</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Divided doses can help address the unique needs of patients who are pregnant</p> <p>In the United States, opioid use by patients who are pregnant more than quadrupled from 1999 to 2014.<sup>1</sup> Opioid use disorder (OUD) in the perinatal period is associated with a higher risk for depression, suicide, malnutrition, domestic violence, and obstetric complications such as spontaneous abortion, preeclampsia, and premature delivery.<sup>2</sup> Buprenorphine and methadone are the standard of care for treating OUD in pregnancy.<sup>3,4</sup> While a literature review found that maternal treatment with buprenorphine has comparable efficacy to treatment with methadone,<sup>5</sup> a small randomized, double-blind study found that compared to buprenorphine, methadone was associated with significantly lower use of additional opioids (<i>P</i> = .047).<sup>6</sup> This suggests methadone has therapeutic value for patients who are pregnant.</p> <p>Despite the benefits of methadone for treating perinatal OUD, the physiological changes that occur in patients who are pregnant—coupled with methadone’s unique pharmacologic properties—may complicate its use. Patients typically take methadone once a day, and the dose is titrated every 3 to 5 days to allow serum levels to reach steady state.<sup>7 </sup>During pregnancy, there are increases in both the volume of distribution and medication metabolism secondary to increased expression of the cytochrome P450 3A4 enzyme by the liver, intestine, and placenta.<sup>8</sup> Additionally, as the pregnancy progresses, the rate of methadone metabolism increases.<sup>9 </sup>Methadone’s half-life (20 to 35 hours) leads to its accumulation in tissue and slow release into the blood.<sup>10</sup> As a result, patients with OUD who are pregnant often require higher doses of methadone or divided dosing, particularly in the second and third trimesters.<sup>11</sup>In this article, we provide a strategy for divided dosing of methadone for managing opioid withdrawal symptoms in the acute care setting. We present 2 cases of women with OUD who are pregnant and describe the collaboration of addiction medicine, consultation-liaison psychiatry, and obstetrics services.</p> <h3> <span class="dingbat"> </span> <span class="box">CASE 1</span> <span class="dingbat"> </span> </h3> <p>Ms. H, age 29, is G3P2 and presents to the emergency department (ED) during her fourth pregnancy at 31 weeks, 1 day gestation. She has a history of opioid, cocaine, and benzodiazepine use disorders and chronic hepatitis C. Ms. H is enrolled in an opioid treatment program and takes methadone 190 mg/d in addition to nonprescribed opioids. In the ED, Ms. H requests medically supervised withdrawal management. Her urine toxicology is positive for cocaine, benzodiazepines, methadone, and opiates. Her laboratory results and electrocardiogram (ECG) are unremarkable. On admission, Ms. H’s Clinical Opiate Withdrawal Scale (COWS) score is 3, indicating minimal symptoms (5 to 12: mild; 13 to 24: moderate; 25 to 36: moderately severe; &gt;36: severe). Fetal monitoring is reassuring.</p> <p>Ms. H’s withdrawal is monitored with COWS every 4 hours. The treatment team initiates methadone 170 mg/d, with an additional 10 mg/d as needed to keep her COWS score &lt;8, and daily QTc monitoring. Ms. H also receives lorazepam 2 to 4 mg/d as needed for benzodiazepine withdrawal. Despite the increase in her daily methadone dose, Ms. H continues to experience opioid withdrawal in the early evening and overnight. As a result, the treatment team increases Ms. H’s morning methadone dose to 190 mg and schedules an afternoon dose of 30 mg. Despite this adjustment, her COWS scores remain elevated in the afternoon and evening, and she requires additional as-needed doses of methadone. Methadone peak and trough levels are ordered to assess for rapid metabolism. The serum trough level is 190 ng/mL, which is low, and a serum peak level is not reported. Despite titration, Ms. H has a self-directed premature discharge.<br/><br/>Five days later at 32 weeks, 2 days gestation, Ms. H is readmitted after she had resumed use of opioids, benzodiazepines, and cocaine. Her vital signs are stable, and her laboratory results and ECG are unremarkable. Fetal monitoring is reassuring. Given Ms. H’s low methadone serum trough level and overall concern for rapid methadone metabolism, the treatment team decides to divide dosing of methadone. Over 9 days, the team titrates methadone to 170 mg twice daily on the day of discharge, which resolves Ms. H’s withdrawal symptoms.<br/><br/>At 38 weeks, 5 days gestation, Ms. H returns to the ED after experiencing labor contractions and opiate withdrawal symptoms after she resumed use of heroin, cocaine, and benzodiazepines. During this admission, Ms. H’s methadone is increased to 180 mg twice daily with additional as-needed doses for ongoing withdrawal symptoms. At 39 weeks, 2 days gestation, Ms. H has a scheduled cesarean delivery.<br/><br/>Her infant has a normal weight but is transferred to the neonatal intensive care unit (NICU) for management of neonatal opioid withdrawal syndrome (NOWS) and receives morphine. The baby remains in the NICU for 35 days and is discharged home without further treatment. When Ms. H is discharged, her methadone dose is 170 mg twice daily, which resolves her opioid withdrawal symptoms. The treatment team directs her to continue care in her methadone outpatient program and receive treatment for her cocaine and benzodiazepine use disorders. She declines residential or inpatient substance use treatment.</p> <p>Ms. M, age 39, is G4P2 and presents to the hospital during her fifth pregnancy at 27 weeks gestation. She has not received prenatal care for this pregnancy. She has a history of OUD and major depressive disorder (MDD). Ms. M’s urine toxicology is positive for opiates, fentanyl, and oxycodone. Her laboratory results are notable for mildly elevated alanine aminotransferase, positive hepatitis C antibody, and a hepatitis C viral load of 91,000, consistent with chronic hepatitis C infection. On admission, her COWS score is 14, indicating moderate withdrawal symptoms. Her ECG is unremarkable, and fetal monitoring is reassuring.</p> <p>Ms. M had received methadone during a prior pregnancy and opts to reinitiate treatment with methadone during her current admission. The team initiates methadone 20 mg/d with additional as-needed doses for ongoing withdrawal symptoms. Due to a persistently elevated COWS score, Ms. M’s methadone is increased to 90 mg/d, which resolves her withdrawal symptoms. However, on Day 4, Ms. M reports having anxiety, refuses bloodwork to obtain methadone peak and trough levels, and prematurely discharges from the hospital. <br/><br/>One day later at 27 weeks, 5 days gestation, Ms. M is readmitted for continued management of opioid withdrawal. She presents with stable vital signs, an unremarkable ECG, and reassuring fetal monitoring. Her COWS score is 5. The treatment team reinitiates methadone at 80 mg/d and titrates it to 100 mg/d on Day 7. Given Ms. M’s ongoing evening cravings and concern for rapid methadone metabolism, on Day 10 the team switches the methadone dosing to 50 mg twice daily to maintain steady-state levels and promote patient comfort. Fluoxetine 20 mg/d is started for comorbid MDD and eventually increased to 80 mg/d. Ms. M is discharged on Day 15 with a regimen of methadone 60 mg/d in the morning and 70 mg/d at night. She plans to resume care in an opioid treatment program and follow up with psychiatry and hepatology for her anxiety and hepatitis C.</p> <h3>A need for aggressive treatment </h3> <p>Given the rising rates of opioid use by patients who are pregnant, harmful behavior related to opioid use, and a wealth of evidence supporting opioid agonist treatment for OUD in pregnancy, there is a growing need for guidance in managing perinatal OUD. A systematic approach to using methadone to treat OUD in patients who are pregnant is essential; the lack of data surrounding use of this medication in such patients may cause overall harm.<sup>12</sup> Limited guidelines and a lack of familiarity with prescribing methadone to patients who are pregnant may lead clinicians to underdose patients, which can result in ongoing withdrawal, premature patient-directed discharges, and poor engagement in care.<sup>13</sup> Both patients in the 2 cases described in this article experienced ongoing withdrawal symptoms despite daily titration of methadone. This suggests rapid metabolism, which was successfully managed by dividing the dosing of methadone, particularly in the latter trimesters.</p> <p>These cases illustrate the need for aggressive perinatal opioid withdrawal management through rapid escalation of divided doses of methadone in a monitored acute care setting. Because methadone elimination is more rapid and clearance rates increase during the perinatal period, divided methadone dosing allows for sustained plasma methadone concentrations and improved outpatient treatment adherence.<sup>9,14,15</sup></p> <h3>Decreasing the rate of premature discharges</h3> <p>In both cases, the patients discharged from the hospital prematurely, likely related to incomplete management of their opioid withdrawal or other withdrawal syndromes (both patients had multiple substance use disorders [SUDs]). Compared to patients without an SUD, patients with SUDs are 3 times more likely to have a self-directed discharge.<sup>16</sup> Patients report leaving the hospital prematurely due to undertreated withdrawal, uncontrolled pain, discrimination by staff, and hospital restrictions.<sup>16</sup> Recommendations to decrease the rates of premature patient-directed discharges in this population include providing patient-centered and harm reduction–oriented care in addition to adequate management of pain and withdrawal.<sup>17</sup></p> <h3>Impact of methadone on fetal outcomes</h3> <p>Approximately 55% to 94% of infants born to patients who are opioid-dependent will develop NOWS. However, there is no relationship between this syndrome and therapeutic doses of methadone.<sup>18</sup> Moreover, long-term research has found that after adjusting for socioeconomic factors, methadone treatment during pregnancy does not have an adverse effect on postnatal development. Divided dosing in maternal methadone administration is also shown to have less of an impact on fetal neurobehavior and NOWS.<sup>19</sup></p> <p>Our recommendations for methadone treatment for perinatal patients are outlined in the <b><i>Table </i></b>(<b><i>page e13</i></b>). Aggressive treatment of opioid withdrawal in the hospital can promote treatment engagement and prevent premature discharges. Clinicians should assess for other withdrawal syndromes when a patient has multiple SUDs and collaborate with an interdisciplinary team to improve patient outcomes.</p> <p> <strong>References</strong> </p> <p class="reference"> 1. Haight SC, Ko JY, Tong VT, et al. Opioid use disorder documented at delivery hospitalization – United States, 1999-2014. MMWR Morb Mortal Wkly Rep. 2018;67(31):845-849.<br/><br/> 2. Kaltenbach K, Berghella V, Finnegan L. Opioid dependence during pregnancy. Effects and management. Obstet Gynecol Clin North Am. 1998;25(1):139-151. doi:10.1016/S0889-8545(05)70362-4<br/><br/> 3. Baumgaertner E. Biden administration offers plan to get addiction-fighting medicine to pregnant women. <span class="iref">The </span><span class="iref">New York Times</span>. October 21, 2022. Accessed February 23, 2023. https://www.nytimes.com/2022/10/21/health/addiction-treatment-pregnancy.html<br/><br/> 4. Jones HE, Fischer G, Heil SH, et al. Maternal Opioid Treatment: Human Experimental Research (MOTHER)--approach, issues and lessons learned. Addiction. 2012;107 Suppl 1(0 1):28-35. doi:10.1111/j.1360-0443.2012.04036.x<br/><br/> 5. Jones HE, Heil SH, Baewert A, et al. Buprenorphine treatment of opioid-dependent pregnant women: a comprehensive review. Addiction. 2012;107 Suppl 1:5-27.<br/><br/> 6. Fischer G, Ortner R, Rohrmeister K, et al. Methadone versus buprenorphine in pregnant addicts: a double-blind, double-dummy comparison study. Addiction. 2006;101(2):275-281. doi:10.1111/j.1360-0443.2006.01321.x<br/><br/> 7. Substance Abuse and Mental Health Services Administration. Chapter 3B: Methadone. <span class="iref">Medications for Opioid Use Disorder: For Healthcare and Addiction Professionals, Policymakers, Patients, and Families: Updated 2021</span>. Substance Abuse and Mental Health Services Administration; August 2021. https://www.ncbi.nlm.nih.gov/books/NBK574918/<br/><br/> 8. Feghali M, Venkataramanan R, Caritis S. Pharmacokinetics of drugs in pregnancy. Semin Perinatol. 2015;39(7):512-519. doi:10.1053/j.semperi.2015.08.003<br/><br/> 9. McCarthy JJ, Vasti EJ, Leamon MH, et al. The use of serum methadone/metabolite ratios to monitor changing perinatal pharmacokinetics. J Addict Med. 2018;12(3): 241-246.<br/><br/> 10. Center for Substance Abuse Treatment. <span class="iref">Medication-Assisted Treatment for Opioid Addiction in Opioid Treatment Programs. Treatment Improvement Protocol Series No. 43.</span> Substance Abuse and Mental Health Service Administration; 2005.<br/><br/> 11. Substance Abuse and Mental Health Services Administration. <span class="iref">Clinical Guidance for Treating Pregnant and Parenting Women with Opioid Use Disorder and Their Infant</span>s. Createspace Independent Publishing Platform; 2018.<br/><br/> 12. Balch B. Prescribing without data: doctors advocate for the inclusion of pregnant people in clinical research. Association of American Medical Colleges. March 22, 2022. Accessed September 30, 2022. https://www.aamc.org/news-insights/prescribing-without-data-doctors-advocate-inclusion-pregnant-people-clinical-research<br/><br/> 13. Leavitt SB. Methadone Dosing &amp; Safety in the Treatment of Opioid Addiction. 2003. <span class="iref">Addiction Treatment Forum</span>. Accessed November 28, 2023. https://atforum.com/documents/DosingandSafetyWP.pdf<br/><br/> 14. McCarthy JJ, Leamon MH, Willitts NH, et al. The effect of methadone dose regimen on neonatal abstinence syndrome. J Addict Med. 2015; 9(2):105-110.<br/><br/> 15. DePetrillo PB, Rice JM. Methadone dosing and pregnancy: impact on program compliance. Int J Addict. 1995;30(2):207-217.<br/><br/> 16. Simon R, Snow R, Wakeman S. Understanding why patients with substance use disorders leave the hospital against medical advice: a qualitative study. Subst Abus. 2020;41(4):519-525. doi:10.1080/08897077.2019.1671942<br/><br/> 17. McNeil R, Small W, Wood E, et al. Hospitals as a ‘risk environment’: an ethno-epidemiological study of voluntary and involuntary discharge from hospital against medical advice among people who inject drugs. Soc Sci Med. 2014;105:59-66.<br/><br/> 18. Jones HE, Jansson LM, O’Grady KE, et al. The relationship between maternal methadone dose at delivery and neonatal outcome: methodological and design considerations. Neurotoxicol Teratol. 2013;39:110-115.<br/><br/> 19. McCarthy JJ, Leamon MH, Parr MS, et al. High-dose methadone maintenance in pregnancy: maternal and neonatal outcomes. Am J Obstet Gynecol. 2005;193(3 Pt 1):606-610.</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
1800267D.SIG
Disable zoom
Off

Delirious mania: Presentation, pathogenesis, and management

Article Type
Changed
Wed, 12/06/2023 - 08:52
Display Headline
Delirious mania: Presentation, pathogenesis, and management

Delirious mania is a syndrome characterized by the acute onset of severe hyperactivity, psychosis, catatonia, and intermittent confusion. While there have been growing reports of this phenomenon over the last 2 decades, it remains poorly recognized and understood.1,2 There is no widely accepted nosology for delirious mania and the condition is absent from DSM-5, which magnifies the difficulties in making a timely diagnosis and initiating appropriate treatment. Delayed diagnosis and treatment may result in a detrimental outcome.2,3 Delirious mania has also been labeled as lethal catatonia, specific febrile delirium, hyperactive or exhaustive mania, and Bell’s mania.2,4,5 The characterization and diagnosis of this condition have a long and inconsistent history (Box1,6-11).

Box

Delirious mania: A brief history

Delirious mania was originally recognized in 1849 by Luther Bell in McLean Hospital after he observed 40 cases that were uniquely distinct from 1,700 other cases from 1836 to 1849.6 He described these patients as being suddenly confused, demonstrating unprovoked combativeness, remarkable decreased need for sleep, excessive motor restlessness, extreme fearfulness, and certain physiological signs, including rapid pulse and sweating. Bell was limited to the psychiatric treatment of his time, which largely was confined to physical restraints. Approximately three-fourths of these patients died.6

Following Bell’s report, this syndrome remained unexplored and rarely described. Some researchers postulated that the development of confusion was a natural progression of late-phase mania in close to 20% of patients.7 However, this did not account for the rapid onset of symptoms as well as certain unexplained movement abnormalities. In 1980, Bond8 presented 3 cases that were similar in nature to Bell’s depiction: acute onset with extraordinary irritability, withdrawal, delirium, and mania.

For the next 2 decades, delirious mania was seldom reported in the literature. The term was often reserved to illustrate when a patient had nothing more than mania with features of delirium.9

By 1996, catatonia became better recognized in its wide array of symptomology and diagnostic scales.10,11 In 1999, in addition to the sudden onset of excitement, paranoia, grandiosity, and disorientation, Fink1 reported catatonic signs including negativism, stereotypy, posturing, grimacing, and echo phenomena in patients with delirious mania. He identified its sensitive response to electroconvulsive therapy.

Delirious mania continues to be met with incertitude in clinical practice, and numerous inconsistencies have been reported in the literature. For example, some cases that have been reported as delirious mania had more evidence of primary delirium due to another medical condition or primary mania.12,13 Other cases have demonstrated swift improvement of symptoms after monotherapy with antipsychotics without a trial of benzodiazepines or electroconvulsive therapy (ECT); the exclusion of a sudden onset questions the validity of the diagnosis and promotes the use of less efficacious treatments.14,15 Other reports have confirmed that the diagnosis is missed when certain symptoms are more predominant, such as a thought disorder (acute schizophrenia), grandiosity and delusional ideation (bipolar disorder [BD]), and less commonly assessed catatonic signs (ambitendency, automatic obedience). These symptoms are mistakenly attributed to the respective disease.1,16 This especially holds true when delirious mania is initially diagnosed as a primary psychosis, which leads to the administration of antipsychotics.17 Other cases have reported that delirious mania was resistant to treatment, but ECT was never pursued.18

In this review, we provide a more comprehensive perspective of the clinical presentation, pathogenesis, and management of delirious mania. We searched PubMed and Google Scholar using the keywords “delirious mania,” “delirious mania AND catatonia,” or “manic delirium.” Most articles we found were case reports, case series, or retrospective chart reviews. There were no systematic reviews, meta analyses, or randomized control trials (RCTs). The 12 articles included in this review consist of 7 individual case reports, 4 case series, and 1 retrospective chart review that describe a total of 36 cases (Table1,2,5,17,19-26).

CP02212026_t1.png

Clinical presentation: What to look for

Patients with delirious mania typically develop symptoms extremely rapidly. In virtually all published literature, symptoms were reported to emerge within hours to days and consisted of severe forms of mania, psychosis, and delirium; 100% of the cases in our review had these symptoms. Commonly reported symptoms were:

  • intense excitement
  • emotional lability
  • grandiose delusions
  • profound insomnia
  • pressured and rapid speech
  • auditory and visual hallucinations
  • hypersexuality
  • thought disorganization.

Exquisite paranoia can also result in violent aggression (and may require the use of physical restraints). Patients may confine themselves to very small spaces (such as a closet) in response to the intense paranoia. Impairments in various neurocognitive domains—including inability to focus; disorientation; language and visuospatial disturbances; difficulty with shifting and sustaining attention; and short-term memory impairments—have been reported. Patients often cannot recall the events during the episode.1,2,5,27,28

Catatonia has been closely associated with delirious mania.29 Features of excited catatonia—such as excessive motor activity, negativism, grimacing, posturing, echolalia, echopraxia, stereotypy, automatic obedience, verbigeration, combativeness, impulsivity, and rigidity—typically accompany delirious mania.1,5,10,19,27

In addition to these symptoms, patients may engage in specific behaviors. They may exhibit inappropriate toileting such as smearing feces on walls or in bags, fecal or urinary incontinence, disrobing or running naked in public places, or pouring liquid on the floor or on one’s head.1,2

Continue to: Of the 36 cases...

 

 

Of the 36 cases reported in the literature we reviewed, 20 (55%) were female. Most patients had an underlining psychiatric condition, including BD (72%), major depressive disorder (8%), and schizophrenia (2%). Three patients had no psychiatric history.

Physical examination

On initial presentation, a patient with delirious mania may be dehydrated, with dry mucous membranes, pale conjunctiva, tongue dryness, and poor skin turgor.28,30 Due to excessive motor activity, diaphoresis with tachycardia, fluctuating blood pressure, and fever may be present.31

Certain basic cognitive tasks should be assessed to determine the patient’s orientation to place, date, and time. Assess if the patient can recall recent events, names of objects, or perform serial 7s; clock drawing capabilities also should be ascertained.1,2,5 A Mini-Mental State Examination is useful.32

The Bush-Francis Catatonia Rating Scale should be used to elicit features of catatonia, such as waxy flexibility, negativism, gegenhalten, mitgehen, catalepsy, ambitendency, automatic obedience, and grasp reflex.10

Laboratory findings are nonspecific

Although no specific laboratory findings are associated with delirious mania, bloodwork and imaging are routinely investigated, especially if delirium characteristics are most striking. A complete blood count, chemistries, hepatic panel, thyroid functioning, blood and/or urine cultures, creatinine phosphokinase (CPK), and urinalysis can be ordered. Head imaging such as MRI and CT to rule out intracranial pathology are typically performed.19 However, the diagnosis of delirious mania is based on the presence of the phenotypic features, by verification of catatonia, and by the responsiveness to the treatment delivered.29

Continue to: Pathogenisis: Several hypotheses

 

 

Pathogenesis: Several hypotheses

The pathogenesis of delirious mania is not well understood. There are several postulations but no salient theory. Most patients with delirious mania have an underlying systemic medical or psychiatric condition.

Mood disorders. Patients with BD or schizoaffective disorder are especially susceptible to delirious mania. The percentage of manic patients who present with delirious mania varies by study. One study suggested approximately 19% have features of the phenomenon,33 while others estimated 15% to 25%.34 Elias et al35 calculated that 15% of patients with mania succumb to manic exhaustion; from this it can be reasonably concluded that these were cases of misdiagnosed delirious mania.

Delirium hypothesis. Patients with delirious mania typically have features of delirium, including fluctuation of consciousness, disorientation, and/or poor sleep-wake cycle.36 During rapid eye movement (REM) and non-REM sleep, memory circuits are fortified. When there is a substantial loss of REM and non-REM sleep, these circuits become faulty, even after 1 night. Pathological brain waves on EEG reflect the inability to reinforce the memory circuits. Patients with these waves may develop hallucinations, bizarre delusions, and altered sensorium. ECT reduces the pathological slow wave morphologies, thus restoring the synaptic maintenance and correcting the incompetent circuitry. This can explain the robust and rapid response of ECT in a patient with delirious mania.37,38

Neurotransmitter hypothesis. It has been shown that in patients with delirious mania there is dysregulation of dopamine transport, which leads to dopamine overflow in the synapse. In contrast to a drug effect (ie, cocaine or methamphetamine) that acts by inhibiting dopamine reuptake, dopamine overflow in delirious mania is caused by the loss of dopamine transporter regulation. This results in a dysfunctional dopaminergic state that precipitates an acute state of delirium and agitation.39,40

Serotonin plays a role in mood disorders, including mania and depression.41,42 More specifically, serotonin has been implicated in impulsivity and aggression as shown by reduced levels of CSF 5-hydroxyindoleacetic acid (5-HIAA) and depletion of 5-hydroxytryptophan (5-HTP).43

Continue to: Alterations in gamma-aminobutyric acid (GABA) transmission...

 

 

Alterations in gamma-aminobutyric acid (GABA) transmission are known to occur in delirium and catatonia. In delirium, GABA signaling is increased, which disrupts the circadian rhythm and melatonin release, thus impairing the sleep-wake cycle.44 Deficiencies in acetylcholine and melatonin are seen as well as excess of other neurotransmitters, including norepinephrine and glutamate.45 Conversely, in catatonia, functional imaging studies found decreased GABA-A binding in orbito­frontal, prefrontal, parietal, and motor cortical regions.46 A study analyzing 10 catatonic patients found decreased density of GABA-A receptors in the left sensorimotor cortex compared to psychiatric and healthy controls.47

Other neurotransmitters, such as glutamate, at the N-methyl-D-aspartate receptors (NMDAR) have been hypothesized to be hyperactive, causing downstream dysregulation of GABA functioning.48 However, the exact connection between delirious mania and all these receptors and neurotransmitters remains unknown.

Encephalitis hypothesis. The relationship between delirious mania and autoimmune encephalitis suggests delirious mania has etiologies other than a primary psychiatric illness. In a 2020 retrospective study49 that analyzed 79 patients with anti-NMDAR encephalitis, 25.3% met criteria for delirious mania, and 95% of these patients had catatonic features. Dalmau et al50 found that in many cases, patients tend to respond to ECT; in a cases series of 3 patients, 2 responded to benzodiazepines.

COVID-19 hypothesis. The SARS-CoV-2 virion has been associated with many neuropsychiatric complications, including mood, psychotic, and neurocognitive disorders.51,52 There also have been cases of COVID-19–induced catatonia.53-55 One case of delirious mania in a patient with COVID-19 has been reported.21 The general mechanism has been proposed to be related to the stimulation of the proinflammatory cytokines, such as tumor necrosis factor-alpha and interleukin-6, which the virus produces in large quantities.56 These cytokines have been linked to psychosis and other psychiatric disorders.57 The patient with COVID-19–induced delirious mania had elevated inflammatory markers, including erythrocyte sedimentation rate, C-reactive protein, ferritin, and D-dimer, which supports a proinflammatory state. This patient had a complete resolution of symptoms with ECT.21

Management: Benzodiazepines and ECT

A step-by-step algorithm for managing delirious mania is outlined in the Figure. Regardless of the underlining etiology, management of delirious mania consists of benzodiazepines (lorazepam and diazepam); prompt use of ECT, particularly for patients who do not improve with large doses of lorazepam; or (if applicable) continued treatment of the underlining medical condition, which does not preclude the use of benzodiazepines or ECT. Recent reports27,58 have described details for using ECT for delirious mania, highlighting the use of high-energy dosing, bilateral electrode placement, and frequent sessions.

CP02212026_f1.png

Continue to: Knowing which medications...

 

 

Knowing which medications to avoid is as important as knowing which agents to administer. Be vigilant in avoiding high-potency antipsychotics, as these medications can worsen extrapyramidal symptoms and may precipitate seizures or neuroleptic malignant syndrome (NMS).28 Anticholinergic agents should also be avoided because they worsen confusion. Although lithium is effective in BD, in delirious mania, high doses of lithium and haloperidol may cause severe encephalopathic syndromes, with symptoms that can include lethargy, tremors, cerebellar dysfunction, and worsened confusion; it may also cause widespread and irreversible brain damage.59While positive outcomes have been documented when using a combination of antipsychotics and lithium,8,60 this approach should be considered carefully and tailored to individual cases, taking into account the severity of manic and psychotic symptoms in addition to the level of catatonia.

Due to long periods of hyperactivity, withdrawal, and diaphoresis, patients with delirious mania may be severely dehydrated with metabolic derangements, including elevated CPK due to rhabdomyolysis from prolonged exertion, IM antipsychotics, or rigidity. To prevent acute renal failure, this must be immediately addressed with rapid fluid resuscitation and electrolyte repletion.61

Benzodiazepines. The rapid use of lorazepam should be initiated when delirious mania is suspected. Doses of 6 to 20 mg have been reported to be effective if tolerated.5,20 Typically, high-dose lorazepam will not have the sedative effect that would normally occur in a patient who does not have delirious mania.2 Lorazepam should be titrated until full resolution of symptoms. Doses up to 30 mg have been reported as effective and tolerable.62 In our literature review, 50% of patients (18/36) responded or partially responded to lorazepam. However, only 3 case reports documented a complete remission with lorazepam, and many patients needed ECT for remission of symptoms.

ECT is generally reserved for patients who are not helped by benzodiazepine therapy, which is estimated to be up to 20%.5 ECT is highly effective in delirious mania, with remission rates ranging from 80% to 100%.1 ECT is also effective in acute non­delirious mania (comparable to depression); however, it is only used in a small minority of cases (0.2% to 12%).35 In our review, 58% of cases (21/36) reported using ECT, and in all cases it resulted in complete remission.

A dramatic improvement can be seen even after a single ECT session, though most patients show improvement after 4 sessions or 3 to 7 days.1,2,5 In our review, most patients received 4 to 12 sessions until achieving complete remission.

Continue to: No RCTs have evaluated...

 

 

No RCTs have evaluated ECT electrode placement in patients with delirious mania. However, several RCTs have investigated electrode placement in patients with acute nondelirious mania. Hiremani et al63 found that bitemporal placement had a more rapid response rate than bifrontal placement, but there was no overall difference in response rate. Barekatain et al64 found no difference between these 2 bilateral approaches. Many of the delirious mania cases report using a bilateral placement (including 42% of the ECT cases in our review) due to the emergent need for rapid relief of symptoms, which is especially necessary if the patient is experiencing hemodynamic instability, excessive violence, risk for self-harm, worsening delirium, or resistance to lorazepam.

Prognosis: Often fatal if left untreated

Patients with delirious mania are at high risk to progress to a more severe form of NMS or malignant catatonia. Therefore, high-potency antipsychotics should be avoided; mortality can be elevated from 60% without antipsychotics to 78% with antipsychotics.4 Some researchers estimate 75% to 78% of cases of delirious mania can be fatal if left untreated.3,6

 

Bottom Line

Delirious mania is routinely mistaken for more conventional manic or psychotic disorders. Clinicians need to be able to rapidly recognize the symptoms of this syndrome, which include mania, psychosis, delirium, and possible catatonia, so they can avoid administering toxic agents and instead initiate effective treatments such as benzodiazepines and electroconvulsive therapy.

Related Resources

Drug Brand Names

Diazepam • Valium
Haloperidol • Haldol
Lithium • Eskalith, Lithobid
Lorazepam • Ativan

[embed:render:related:node:249909]

References

1. Fink M. Delirious mania. Bipolar Disord. 1999;1(1):54-60.

2. Karmacharya R, England ML, Ongür D. Delirious mania: clinical features and treatment response. J Affect Disord. 2008;109(3):312-316.

3. Friedman RS, Mufson MJ, Eisenberg TD, et al. Medically and psychiatrically ill: the challenge of delirious mania. Harv Rev Psychiatry. 2003;11(2):91-98.

4. Mann SC, Caroff SN, Bleier HR, et al. Lethal catatonia. Am J Psychiatry. 1986;143(11):1374-1381.

5. Detweiler MB, Mehra A, Rowell T, et al. Delirious mania and malignant catatonia: a report of 3 cases and review. Psychiatr Q. 2009;80(1):23-40.

6. Bell L. On a form of disease resembling some advanced stages of mania and fever. American Journal of Insanity. 1849;6(2):97-127.

7. Carlson GA, Goodwin FK. The stages of mania. A longitudinal analysis of the manic episode. Arch Gen Psychiatry. 1973;28(2):221-228.

8. Bond TC. Recognition of acute delirious mania. Arch Gen Psychiatry. 1980;37(5):553-554.

9. Hutchinson G, David A. Manic pseudo-delirium - two case reports. Behav Neurol. 1997;10(1):21-23.

10. Bush G, Fink M, Petrides G, et al. Catatonia. I. Rating scale and standardized examination. Acta Psychiatr Scand. 1996;93(2):129-136.

11. Bush G, Fink M, Petrides G, et al. Catatonia. II. Treatment with lorazepam and electroconvulsive therapy. Acta Psychiatr Scand. 1996;93(2):137-143.

12. Cordeiro CR, Saraiva R, Côrte-Real B, et al. When the bell rings: clinical features of Bell’s mania. Prim Care Companion CNS Disord. 2020;22(2):19l02511. doi:10.4088/PCC.19l02511

13. Yeo LX, Kuo TC, Hu KC, et al. Lurasidone-induced delirious mania. Am J Ther. 2019;26(6):e786-e787.

14. Jung WY, Lee BD. Quetiapine treatment for delirious mania in a military soldier. Prim Care Companion J Clin Psychiatry. 2010;12(2):PCC.09l00830. doi:10.4088/PCC.09l00830yel

15. Wahid N, Chin G, Turner AH, et al. Clinical response of clozapine as a treatment for delirious mania. Ment Illn. 2017;9(2):7182. doi:10.4081/mi.2017.7182

16. Taylor MA, Fink M. Catatonia in psychiatric classification: a home of its own. Am J Psychiatry. 2003;160(7):1233-1241.

17. Danivas V, Behere RV, Varambally S, et al. Electroconvulsive therapy in the treatment of delirious mania: a report of 2 patients. J ECT. 2010;26(4):278-279.

18. O’Callaghan N, McDonald C, Hallahan B. Delirious mania intractable to treatment. Ir J Psychol Med. 2016;33(2):129-132.

19. Vasudev K, Grunze H. What works for delirious catatonic mania? BMJ Case Rep. 2010;2010:bcr0220102713. doi:10.1136/bcr.02.2010.2713

20. Jacobowski NL, Heckers S, Bobo WV. Delirious mania: detection, diagnosis, and clinical management in the acute setting. J Psychiatr Pract. 2013;19(1):15-28.

21. Reinfeld S, Yacoub A. A case of delirious mania induced by COVID-19 treated with electroconvulsive therapy. J ECT. 2021;37(4):e38-e39.

22. Lee BS, Huang SS, Hsu WY, et al. Clinical features of delirious mania: a series of five cases and a brief literature review. BMC Psychiatry. 2012;12:65. doi:10.1186/1471-244X-12-65

23. Bipeta R, Khan MA. Delirious mania: can we get away with this concept? A case report and review of the literature. Case Rep Psychiatry. 2012;2012:720354. doi:10.1155/2012/720354

24. Nunes AL, Cheniaux E. Delirium and mania with catatonic features in a Brazilian patient: response to ECT. J Neuropsychiatry Clin Neurosci. 2014;26(1):E1-E3.

25. Tegin C, Kalayil G, Lippmann S. Electroconvulsive therapy and delirious catatonic mania. J ECT. 2017;33(4):e33-e34.

26. Melo AL, Serra M. Delirious mania and catatonia. Bipolar Disord. 2020;22(6):647-649.

27. Fink M. Expanding the catatonia tent: recognizing electroconvulsive therapy responsive syndromes. J ECT. 2021;37(2):77-79.

28. Fink M. Electroconvulsive Therapy: A Guide for Professionals and Their Patients. Oxford University Press; 2009.

29. Fink M, Taylor MA. The many varieties of catatonia. Eur Arch Psychiatry Clin Neurosci. 2001;251 Suppl 1:I8-I13.

30. Vivanti A, Harvey K, Ash S, et al. Clinical assessment of dehydration in older people admitted to hospital: what are the strongest indicators? Arch Gerontol Geriatr. 2008;47(3):340-355.

31. Ware MR, Feller DB, Hall KL. Neuroleptic malignant syndrome: diagnosis and management. Prim Care Companion CNS Disord. 2018;20(1):17r02185. doi:10.4088/PCC.17r0218

32. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189-198.

33. Taylor MA, Abrams R. The phenomenology of mania. A new look at some old patients. Arch Gen Psychiatry. 1973;29(4):520-522.

34. Klerman GL. The spectrum of mania. Compr Psychiatry. 1981;22(1):11-20.

35. Elias A, Thomas N, Sackeim HA. Electroconvulsive therapy in mania: a review of 80 years of clinical experience. Am J Psychiatry. 2021;178(3):229-239.

36. Thom RP, Levy-Carrick NC, Bui M, et al. Delirium. Am J Psychiatry. 2019;176(10):785-793.

37. Charlton BG, Kavanau JL. Delirium and psychotic symptoms--an integrative model. Med Hypotheses. 2002;58(1):24-27.

38. Kramp P, Bolwig TG. Electroconvulsive therapy in acute delirious states. Compr Psychiatry. 1981;22(4):368-371.

39. Mash DC. Excited delirium and sudden death: a syndromal disorder at the extreme end of the neuropsychiatric continuum. Front Physiol. 2016;7:435.

40. Strawn JR, Keck PE Jr, Caroff SN. Neuroleptic malignant syndrome. Am J Psychiatry. 2007;164(6):870-876.

41. Charney DS. Monoamine dysfunction and the pathophysiology and treatment of depression. J Clin Psychiatry. 1998;59 Suppl 14:11-14.

42. Shiah IS, Yatham LN. Serotonin in mania and in the mechanism of action of mood stabilizers: a review of clinical studies. Bipolar Disord. 2000;2(2):77-92.

43. Dalley JW, Roiser JP. Dopamine, serotonin and impulsivity. Neuroscience. 2012;215:42-58.

44. Maldonado JR. Pathoetiological model of delirium: a comprehensive understanding of the neurobiology of delirium and an evidence-based approach to prevention and treatment. Crit Care Clin. 2008;24(4):789-856, ix.

45. Maldonado JR. Neuropathogenesis of delirium: review of current etiologic theories and common pathways. Am J Geriatr Psychiatry. 2013;21(12):1190-1222.

46. Rasmussen SA, Mazurek MF, Rosebush PI. Catatonia: our current understanding of its diagnosis, treatment and pathophysiology. World J Psychiatry. 2016;6(4):391-398.

47. Northoff G, Steinke R, Czcervenka C, et al. Decreased density of GABA-A receptors in the left sensorimotor cortex in akinetic catatonia: investigation of in vivo benzodiazepine receptor binding. J Neurol Neurosurg Psychiatry. 1999;67(4):445-450.

48. Daniels J. Catatonia: clinical aspects and neurobiological correlates. J Neuropsychiatry Clin Neurosci. 2009;21(4):371-380.

49. Restrepo-Martínez M, Chacón-González J, Bayliss L, et al. Delirious mania as a neuropsychiatric presentation in patients with anti-N-methyl-D-aspartate receptor encephalitis. Psychosomatics. 2020;61(1):64-69.

50. Dalmau J, Armangué T, Planagumà J, et al. An update on anti-NMDA receptor encephalitis for neurologists and psychiatrists: mechanisms and models. Lancet Neurol. 2019;18(11):1045-1057.

51. Steardo L Jr, Steardo L, Verkhratsky A. Psychiatric face of COVID-19. Transl Psychiatry. 2020;10(1):261.

52. Iqbal Y, Al Abdulla MA, Albrahim S, et al. Psychiatric presentation of patients with acute SARS-CoV-2 infection: a retrospective review of 50 consecutive patients seen by a consultation-liaison psychiatry team. BJPsych Open. 2020;6(5):e109.

53. Gouse BM, Spears WE, Nieves Archibald A, et al. Catatonia in a hospitalized patient with COVID-19 and proposed immune-mediated mechanism. Brain Behav Immun. 2020;89:529-530.

54. Caan MP, Lim CT, Howard M. A case of catatonia in a man with COVID-19. Psychosomatics. 2020;61(5):556-560.

55. Zain SM, Muthukanagaraj P, Rahman N. Excited catatonia - a delayed neuropsychiatric complication of COVID-19 infection. Cureus. 2021;13(3):e13891.

56. Chowdhury MA, Hossain N, Kashem MA, et al. Immune response in COVID-19: a review. J Infect Public Health. 2020;13(11):1619-1629.

57. Radhakrishnan R, Kaser M, Guloksuz S. The link between the immune system, environment, and psychosis. Schizophr Bull. 2017;43(4):693-697.

58. Fink M, Kellner CH, McCall WV. Optimizing ECT technique in treating catatonia. J ECT. 2016;32(3):149-150.

59. Cohen WJ, Cohen NH. Lithium carbonate, haloperidol, and irreversible brain damage. JAMA. 1974;230(9):1283-1287.

60. Davis MJ, de Nesnera A, Folks DG. Confused and nearly naked after going on spending sprees. Current Psychiatry. 2014;13(7):56-62.

61. Stanley M, Chippa V, Aeddula NR, et al. Rhabdomyolysis. StatPearls Publishing; 2021.

62. Fink M, Taylor MA. The catatonia syndrome: forgotten but not gone. Arch Gen Psychiatry. 2009;66(11):1173-1177.

63. Hiremani RM, Thirthalli J, Tharayil BS, et al. Double-blind randomized controlled study comparing short-term efficacy of bifrontal and bitemporal electroconvulsive therapy in acute mania. Bipolar Disord. 2008;10(6):701-707.

64. Barekatain M, Jahangard L, Haghighi M, et al. Bifrontal versus bitemporal electroconvulsive therapy in severe manic patients. J ECT. 2008;24(3):199-202.

Article PDF
Author and Disclosure Information

Samuel Reinfeld, DO
Adeeb Yacoub, MD

• • • •

Department of Psychiatry and Behavioral Health Stony Brook University Renaissance School of Medicine
Stony Brook, New York

Acknowledgments
The authors express great gratitude to Professor Max Fink for his characterization of delirious mania. We also thank Dr. Fink for his continued guidance and support during the preparation of this manuscript.

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Issue
Current Psychiatry - 22(12)
Publications
Topics
Page Number
26-34,54
Sections
Author and Disclosure Information

Samuel Reinfeld, DO
Adeeb Yacoub, MD

• • • •

Department of Psychiatry and Behavioral Health Stony Brook University Renaissance School of Medicine
Stony Brook, New York

Acknowledgments
The authors express great gratitude to Professor Max Fink for his characterization of delirious mania. We also thank Dr. Fink for his continued guidance and support during the preparation of this manuscript.

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Author and Disclosure Information

Samuel Reinfeld, DO
Adeeb Yacoub, MD

• • • •

Department of Psychiatry and Behavioral Health Stony Brook University Renaissance School of Medicine
Stony Brook, New York

Acknowledgments
The authors express great gratitude to Professor Max Fink for his characterization of delirious mania. We also thank Dr. Fink for his continued guidance and support during the preparation of this manuscript.

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Article PDF
Article PDF

Delirious mania is a syndrome characterized by the acute onset of severe hyperactivity, psychosis, catatonia, and intermittent confusion. While there have been growing reports of this phenomenon over the last 2 decades, it remains poorly recognized and understood.1,2 There is no widely accepted nosology for delirious mania and the condition is absent from DSM-5, which magnifies the difficulties in making a timely diagnosis and initiating appropriate treatment. Delayed diagnosis and treatment may result in a detrimental outcome.2,3 Delirious mania has also been labeled as lethal catatonia, specific febrile delirium, hyperactive or exhaustive mania, and Bell’s mania.2,4,5 The characterization and diagnosis of this condition have a long and inconsistent history (Box1,6-11).

Box

Delirious mania: A brief history

Delirious mania was originally recognized in 1849 by Luther Bell in McLean Hospital after he observed 40 cases that were uniquely distinct from 1,700 other cases from 1836 to 1849.6 He described these patients as being suddenly confused, demonstrating unprovoked combativeness, remarkable decreased need for sleep, excessive motor restlessness, extreme fearfulness, and certain physiological signs, including rapid pulse and sweating. Bell was limited to the psychiatric treatment of his time, which largely was confined to physical restraints. Approximately three-fourths of these patients died.6

Following Bell’s report, this syndrome remained unexplored and rarely described. Some researchers postulated that the development of confusion was a natural progression of late-phase mania in close to 20% of patients.7 However, this did not account for the rapid onset of symptoms as well as certain unexplained movement abnormalities. In 1980, Bond8 presented 3 cases that were similar in nature to Bell’s depiction: acute onset with extraordinary irritability, withdrawal, delirium, and mania.

For the next 2 decades, delirious mania was seldom reported in the literature. The term was often reserved to illustrate when a patient had nothing more than mania with features of delirium.9

By 1996, catatonia became better recognized in its wide array of symptomology and diagnostic scales.10,11 In 1999, in addition to the sudden onset of excitement, paranoia, grandiosity, and disorientation, Fink1 reported catatonic signs including negativism, stereotypy, posturing, grimacing, and echo phenomena in patients with delirious mania. He identified its sensitive response to electroconvulsive therapy.

Delirious mania continues to be met with incertitude in clinical practice, and numerous inconsistencies have been reported in the literature. For example, some cases that have been reported as delirious mania had more evidence of primary delirium due to another medical condition or primary mania.12,13 Other cases have demonstrated swift improvement of symptoms after monotherapy with antipsychotics without a trial of benzodiazepines or electroconvulsive therapy (ECT); the exclusion of a sudden onset questions the validity of the diagnosis and promotes the use of less efficacious treatments.14,15 Other reports have confirmed that the diagnosis is missed when certain symptoms are more predominant, such as a thought disorder (acute schizophrenia), grandiosity and delusional ideation (bipolar disorder [BD]), and less commonly assessed catatonic signs (ambitendency, automatic obedience). These symptoms are mistakenly attributed to the respective disease.1,16 This especially holds true when delirious mania is initially diagnosed as a primary psychosis, which leads to the administration of antipsychotics.17 Other cases have reported that delirious mania was resistant to treatment, but ECT was never pursued.18

In this review, we provide a more comprehensive perspective of the clinical presentation, pathogenesis, and management of delirious mania. We searched PubMed and Google Scholar using the keywords “delirious mania,” “delirious mania AND catatonia,” or “manic delirium.” Most articles we found were case reports, case series, or retrospective chart reviews. There were no systematic reviews, meta analyses, or randomized control trials (RCTs). The 12 articles included in this review consist of 7 individual case reports, 4 case series, and 1 retrospective chart review that describe a total of 36 cases (Table1,2,5,17,19-26).

CP02212026_t1.png

Clinical presentation: What to look for

Patients with delirious mania typically develop symptoms extremely rapidly. In virtually all published literature, symptoms were reported to emerge within hours to days and consisted of severe forms of mania, psychosis, and delirium; 100% of the cases in our review had these symptoms. Commonly reported symptoms were:

  • intense excitement
  • emotional lability
  • grandiose delusions
  • profound insomnia
  • pressured and rapid speech
  • auditory and visual hallucinations
  • hypersexuality
  • thought disorganization.

Exquisite paranoia can also result in violent aggression (and may require the use of physical restraints). Patients may confine themselves to very small spaces (such as a closet) in response to the intense paranoia. Impairments in various neurocognitive domains—including inability to focus; disorientation; language and visuospatial disturbances; difficulty with shifting and sustaining attention; and short-term memory impairments—have been reported. Patients often cannot recall the events during the episode.1,2,5,27,28

Catatonia has been closely associated with delirious mania.29 Features of excited catatonia—such as excessive motor activity, negativism, grimacing, posturing, echolalia, echopraxia, stereotypy, automatic obedience, verbigeration, combativeness, impulsivity, and rigidity—typically accompany delirious mania.1,5,10,19,27

In addition to these symptoms, patients may engage in specific behaviors. They may exhibit inappropriate toileting such as smearing feces on walls or in bags, fecal or urinary incontinence, disrobing or running naked in public places, or pouring liquid on the floor or on one’s head.1,2

Continue to: Of the 36 cases...

 

 

Of the 36 cases reported in the literature we reviewed, 20 (55%) were female. Most patients had an underlining psychiatric condition, including BD (72%), major depressive disorder (8%), and schizophrenia (2%). Three patients had no psychiatric history.

Physical examination

On initial presentation, a patient with delirious mania may be dehydrated, with dry mucous membranes, pale conjunctiva, tongue dryness, and poor skin turgor.28,30 Due to excessive motor activity, diaphoresis with tachycardia, fluctuating blood pressure, and fever may be present.31

Certain basic cognitive tasks should be assessed to determine the patient’s orientation to place, date, and time. Assess if the patient can recall recent events, names of objects, or perform serial 7s; clock drawing capabilities also should be ascertained.1,2,5 A Mini-Mental State Examination is useful.32

The Bush-Francis Catatonia Rating Scale should be used to elicit features of catatonia, such as waxy flexibility, negativism, gegenhalten, mitgehen, catalepsy, ambitendency, automatic obedience, and grasp reflex.10

Laboratory findings are nonspecific

Although no specific laboratory findings are associated with delirious mania, bloodwork and imaging are routinely investigated, especially if delirium characteristics are most striking. A complete blood count, chemistries, hepatic panel, thyroid functioning, blood and/or urine cultures, creatinine phosphokinase (CPK), and urinalysis can be ordered. Head imaging such as MRI and CT to rule out intracranial pathology are typically performed.19 However, the diagnosis of delirious mania is based on the presence of the phenotypic features, by verification of catatonia, and by the responsiveness to the treatment delivered.29

Continue to: Pathogenisis: Several hypotheses

 

 

Pathogenesis: Several hypotheses

The pathogenesis of delirious mania is not well understood. There are several postulations but no salient theory. Most patients with delirious mania have an underlying systemic medical or psychiatric condition.

Mood disorders. Patients with BD or schizoaffective disorder are especially susceptible to delirious mania. The percentage of manic patients who present with delirious mania varies by study. One study suggested approximately 19% have features of the phenomenon,33 while others estimated 15% to 25%.34 Elias et al35 calculated that 15% of patients with mania succumb to manic exhaustion; from this it can be reasonably concluded that these were cases of misdiagnosed delirious mania.

Delirium hypothesis. Patients with delirious mania typically have features of delirium, including fluctuation of consciousness, disorientation, and/or poor sleep-wake cycle.36 During rapid eye movement (REM) and non-REM sleep, memory circuits are fortified. When there is a substantial loss of REM and non-REM sleep, these circuits become faulty, even after 1 night. Pathological brain waves on EEG reflect the inability to reinforce the memory circuits. Patients with these waves may develop hallucinations, bizarre delusions, and altered sensorium. ECT reduces the pathological slow wave morphologies, thus restoring the synaptic maintenance and correcting the incompetent circuitry. This can explain the robust and rapid response of ECT in a patient with delirious mania.37,38

Neurotransmitter hypothesis. It has been shown that in patients with delirious mania there is dysregulation of dopamine transport, which leads to dopamine overflow in the synapse. In contrast to a drug effect (ie, cocaine or methamphetamine) that acts by inhibiting dopamine reuptake, dopamine overflow in delirious mania is caused by the loss of dopamine transporter regulation. This results in a dysfunctional dopaminergic state that precipitates an acute state of delirium and agitation.39,40

Serotonin plays a role in mood disorders, including mania and depression.41,42 More specifically, serotonin has been implicated in impulsivity and aggression as shown by reduced levels of CSF 5-hydroxyindoleacetic acid (5-HIAA) and depletion of 5-hydroxytryptophan (5-HTP).43

Continue to: Alterations in gamma-aminobutyric acid (GABA) transmission...

 

 

Alterations in gamma-aminobutyric acid (GABA) transmission are known to occur in delirium and catatonia. In delirium, GABA signaling is increased, which disrupts the circadian rhythm and melatonin release, thus impairing the sleep-wake cycle.44 Deficiencies in acetylcholine and melatonin are seen as well as excess of other neurotransmitters, including norepinephrine and glutamate.45 Conversely, in catatonia, functional imaging studies found decreased GABA-A binding in orbito­frontal, prefrontal, parietal, and motor cortical regions.46 A study analyzing 10 catatonic patients found decreased density of GABA-A receptors in the left sensorimotor cortex compared to psychiatric and healthy controls.47

Other neurotransmitters, such as glutamate, at the N-methyl-D-aspartate receptors (NMDAR) have been hypothesized to be hyperactive, causing downstream dysregulation of GABA functioning.48 However, the exact connection between delirious mania and all these receptors and neurotransmitters remains unknown.

Encephalitis hypothesis. The relationship between delirious mania and autoimmune encephalitis suggests delirious mania has etiologies other than a primary psychiatric illness. In a 2020 retrospective study49 that analyzed 79 patients with anti-NMDAR encephalitis, 25.3% met criteria for delirious mania, and 95% of these patients had catatonic features. Dalmau et al50 found that in many cases, patients tend to respond to ECT; in a cases series of 3 patients, 2 responded to benzodiazepines.

COVID-19 hypothesis. The SARS-CoV-2 virion has been associated with many neuropsychiatric complications, including mood, psychotic, and neurocognitive disorders.51,52 There also have been cases of COVID-19–induced catatonia.53-55 One case of delirious mania in a patient with COVID-19 has been reported.21 The general mechanism has been proposed to be related to the stimulation of the proinflammatory cytokines, such as tumor necrosis factor-alpha and interleukin-6, which the virus produces in large quantities.56 These cytokines have been linked to psychosis and other psychiatric disorders.57 The patient with COVID-19–induced delirious mania had elevated inflammatory markers, including erythrocyte sedimentation rate, C-reactive protein, ferritin, and D-dimer, which supports a proinflammatory state. This patient had a complete resolution of symptoms with ECT.21

Management: Benzodiazepines and ECT

A step-by-step algorithm for managing delirious mania is outlined in the Figure. Regardless of the underlining etiology, management of delirious mania consists of benzodiazepines (lorazepam and diazepam); prompt use of ECT, particularly for patients who do not improve with large doses of lorazepam; or (if applicable) continued treatment of the underlining medical condition, which does not preclude the use of benzodiazepines or ECT. Recent reports27,58 have described details for using ECT for delirious mania, highlighting the use of high-energy dosing, bilateral electrode placement, and frequent sessions.

CP02212026_f1.png

Continue to: Knowing which medications...

 

 

Knowing which medications to avoid is as important as knowing which agents to administer. Be vigilant in avoiding high-potency antipsychotics, as these medications can worsen extrapyramidal symptoms and may precipitate seizures or neuroleptic malignant syndrome (NMS).28 Anticholinergic agents should also be avoided because they worsen confusion. Although lithium is effective in BD, in delirious mania, high doses of lithium and haloperidol may cause severe encephalopathic syndromes, with symptoms that can include lethargy, tremors, cerebellar dysfunction, and worsened confusion; it may also cause widespread and irreversible brain damage.59While positive outcomes have been documented when using a combination of antipsychotics and lithium,8,60 this approach should be considered carefully and tailored to individual cases, taking into account the severity of manic and psychotic symptoms in addition to the level of catatonia.

Due to long periods of hyperactivity, withdrawal, and diaphoresis, patients with delirious mania may be severely dehydrated with metabolic derangements, including elevated CPK due to rhabdomyolysis from prolonged exertion, IM antipsychotics, or rigidity. To prevent acute renal failure, this must be immediately addressed with rapid fluid resuscitation and electrolyte repletion.61

Benzodiazepines. The rapid use of lorazepam should be initiated when delirious mania is suspected. Doses of 6 to 20 mg have been reported to be effective if tolerated.5,20 Typically, high-dose lorazepam will not have the sedative effect that would normally occur in a patient who does not have delirious mania.2 Lorazepam should be titrated until full resolution of symptoms. Doses up to 30 mg have been reported as effective and tolerable.62 In our literature review, 50% of patients (18/36) responded or partially responded to lorazepam. However, only 3 case reports documented a complete remission with lorazepam, and many patients needed ECT for remission of symptoms.

ECT is generally reserved for patients who are not helped by benzodiazepine therapy, which is estimated to be up to 20%.5 ECT is highly effective in delirious mania, with remission rates ranging from 80% to 100%.1 ECT is also effective in acute non­delirious mania (comparable to depression); however, it is only used in a small minority of cases (0.2% to 12%).35 In our review, 58% of cases (21/36) reported using ECT, and in all cases it resulted in complete remission.

A dramatic improvement can be seen even after a single ECT session, though most patients show improvement after 4 sessions or 3 to 7 days.1,2,5 In our review, most patients received 4 to 12 sessions until achieving complete remission.

Continue to: No RCTs have evaluated...

 

 

No RCTs have evaluated ECT electrode placement in patients with delirious mania. However, several RCTs have investigated electrode placement in patients with acute nondelirious mania. Hiremani et al63 found that bitemporal placement had a more rapid response rate than bifrontal placement, but there was no overall difference in response rate. Barekatain et al64 found no difference between these 2 bilateral approaches. Many of the delirious mania cases report using a bilateral placement (including 42% of the ECT cases in our review) due to the emergent need for rapid relief of symptoms, which is especially necessary if the patient is experiencing hemodynamic instability, excessive violence, risk for self-harm, worsening delirium, or resistance to lorazepam.

Prognosis: Often fatal if left untreated

Patients with delirious mania are at high risk to progress to a more severe form of NMS or malignant catatonia. Therefore, high-potency antipsychotics should be avoided; mortality can be elevated from 60% without antipsychotics to 78% with antipsychotics.4 Some researchers estimate 75% to 78% of cases of delirious mania can be fatal if left untreated.3,6

 

Bottom Line

Delirious mania is routinely mistaken for more conventional manic or psychotic disorders. Clinicians need to be able to rapidly recognize the symptoms of this syndrome, which include mania, psychosis, delirium, and possible catatonia, so they can avoid administering toxic agents and instead initiate effective treatments such as benzodiazepines and electroconvulsive therapy.

Related Resources

Drug Brand Names

Diazepam • Valium
Haloperidol • Haldol
Lithium • Eskalith, Lithobid
Lorazepam • Ativan

[embed:render:related:node:249909]

Delirious mania is a syndrome characterized by the acute onset of severe hyperactivity, psychosis, catatonia, and intermittent confusion. While there have been growing reports of this phenomenon over the last 2 decades, it remains poorly recognized and understood.1,2 There is no widely accepted nosology for delirious mania and the condition is absent from DSM-5, which magnifies the difficulties in making a timely diagnosis and initiating appropriate treatment. Delayed diagnosis and treatment may result in a detrimental outcome.2,3 Delirious mania has also been labeled as lethal catatonia, specific febrile delirium, hyperactive or exhaustive mania, and Bell’s mania.2,4,5 The characterization and diagnosis of this condition have a long and inconsistent history (Box1,6-11).

Box

Delirious mania: A brief history

Delirious mania was originally recognized in 1849 by Luther Bell in McLean Hospital after he observed 40 cases that were uniquely distinct from 1,700 other cases from 1836 to 1849.6 He described these patients as being suddenly confused, demonstrating unprovoked combativeness, remarkable decreased need for sleep, excessive motor restlessness, extreme fearfulness, and certain physiological signs, including rapid pulse and sweating. Bell was limited to the psychiatric treatment of his time, which largely was confined to physical restraints. Approximately three-fourths of these patients died.6

Following Bell’s report, this syndrome remained unexplored and rarely described. Some researchers postulated that the development of confusion was a natural progression of late-phase mania in close to 20% of patients.7 However, this did not account for the rapid onset of symptoms as well as certain unexplained movement abnormalities. In 1980, Bond8 presented 3 cases that were similar in nature to Bell’s depiction: acute onset with extraordinary irritability, withdrawal, delirium, and mania.

For the next 2 decades, delirious mania was seldom reported in the literature. The term was often reserved to illustrate when a patient had nothing more than mania with features of delirium.9

By 1996, catatonia became better recognized in its wide array of symptomology and diagnostic scales.10,11 In 1999, in addition to the sudden onset of excitement, paranoia, grandiosity, and disorientation, Fink1 reported catatonic signs including negativism, stereotypy, posturing, grimacing, and echo phenomena in patients with delirious mania. He identified its sensitive response to electroconvulsive therapy.

Delirious mania continues to be met with incertitude in clinical practice, and numerous inconsistencies have been reported in the literature. For example, some cases that have been reported as delirious mania had more evidence of primary delirium due to another medical condition or primary mania.12,13 Other cases have demonstrated swift improvement of symptoms after monotherapy with antipsychotics without a trial of benzodiazepines or electroconvulsive therapy (ECT); the exclusion of a sudden onset questions the validity of the diagnosis and promotes the use of less efficacious treatments.14,15 Other reports have confirmed that the diagnosis is missed when certain symptoms are more predominant, such as a thought disorder (acute schizophrenia), grandiosity and delusional ideation (bipolar disorder [BD]), and less commonly assessed catatonic signs (ambitendency, automatic obedience). These symptoms are mistakenly attributed to the respective disease.1,16 This especially holds true when delirious mania is initially diagnosed as a primary psychosis, which leads to the administration of antipsychotics.17 Other cases have reported that delirious mania was resistant to treatment, but ECT was never pursued.18

In this review, we provide a more comprehensive perspective of the clinical presentation, pathogenesis, and management of delirious mania. We searched PubMed and Google Scholar using the keywords “delirious mania,” “delirious mania AND catatonia,” or “manic delirium.” Most articles we found were case reports, case series, or retrospective chart reviews. There were no systematic reviews, meta analyses, or randomized control trials (RCTs). The 12 articles included in this review consist of 7 individual case reports, 4 case series, and 1 retrospective chart review that describe a total of 36 cases (Table1,2,5,17,19-26).

CP02212026_t1.png

Clinical presentation: What to look for

Patients with delirious mania typically develop symptoms extremely rapidly. In virtually all published literature, symptoms were reported to emerge within hours to days and consisted of severe forms of mania, psychosis, and delirium; 100% of the cases in our review had these symptoms. Commonly reported symptoms were:

  • intense excitement
  • emotional lability
  • grandiose delusions
  • profound insomnia
  • pressured and rapid speech
  • auditory and visual hallucinations
  • hypersexuality
  • thought disorganization.

Exquisite paranoia can also result in violent aggression (and may require the use of physical restraints). Patients may confine themselves to very small spaces (such as a closet) in response to the intense paranoia. Impairments in various neurocognitive domains—including inability to focus; disorientation; language and visuospatial disturbances; difficulty with shifting and sustaining attention; and short-term memory impairments—have been reported. Patients often cannot recall the events during the episode.1,2,5,27,28

Catatonia has been closely associated with delirious mania.29 Features of excited catatonia—such as excessive motor activity, negativism, grimacing, posturing, echolalia, echopraxia, stereotypy, automatic obedience, verbigeration, combativeness, impulsivity, and rigidity—typically accompany delirious mania.1,5,10,19,27

In addition to these symptoms, patients may engage in specific behaviors. They may exhibit inappropriate toileting such as smearing feces on walls or in bags, fecal or urinary incontinence, disrobing or running naked in public places, or pouring liquid on the floor or on one’s head.1,2

Continue to: Of the 36 cases...

 

 

Of the 36 cases reported in the literature we reviewed, 20 (55%) were female. Most patients had an underlining psychiatric condition, including BD (72%), major depressive disorder (8%), and schizophrenia (2%). Three patients had no psychiatric history.

Physical examination

On initial presentation, a patient with delirious mania may be dehydrated, with dry mucous membranes, pale conjunctiva, tongue dryness, and poor skin turgor.28,30 Due to excessive motor activity, diaphoresis with tachycardia, fluctuating blood pressure, and fever may be present.31

Certain basic cognitive tasks should be assessed to determine the patient’s orientation to place, date, and time. Assess if the patient can recall recent events, names of objects, or perform serial 7s; clock drawing capabilities also should be ascertained.1,2,5 A Mini-Mental State Examination is useful.32

The Bush-Francis Catatonia Rating Scale should be used to elicit features of catatonia, such as waxy flexibility, negativism, gegenhalten, mitgehen, catalepsy, ambitendency, automatic obedience, and grasp reflex.10

Laboratory findings are nonspecific

Although no specific laboratory findings are associated with delirious mania, bloodwork and imaging are routinely investigated, especially if delirium characteristics are most striking. A complete blood count, chemistries, hepatic panel, thyroid functioning, blood and/or urine cultures, creatinine phosphokinase (CPK), and urinalysis can be ordered. Head imaging such as MRI and CT to rule out intracranial pathology are typically performed.19 However, the diagnosis of delirious mania is based on the presence of the phenotypic features, by verification of catatonia, and by the responsiveness to the treatment delivered.29

Continue to: Pathogenisis: Several hypotheses

 

 

Pathogenesis: Several hypotheses

The pathogenesis of delirious mania is not well understood. There are several postulations but no salient theory. Most patients with delirious mania have an underlying systemic medical or psychiatric condition.

Mood disorders. Patients with BD or schizoaffective disorder are especially susceptible to delirious mania. The percentage of manic patients who present with delirious mania varies by study. One study suggested approximately 19% have features of the phenomenon,33 while others estimated 15% to 25%.34 Elias et al35 calculated that 15% of patients with mania succumb to manic exhaustion; from this it can be reasonably concluded that these were cases of misdiagnosed delirious mania.

Delirium hypothesis. Patients with delirious mania typically have features of delirium, including fluctuation of consciousness, disorientation, and/or poor sleep-wake cycle.36 During rapid eye movement (REM) and non-REM sleep, memory circuits are fortified. When there is a substantial loss of REM and non-REM sleep, these circuits become faulty, even after 1 night. Pathological brain waves on EEG reflect the inability to reinforce the memory circuits. Patients with these waves may develop hallucinations, bizarre delusions, and altered sensorium. ECT reduces the pathological slow wave morphologies, thus restoring the synaptic maintenance and correcting the incompetent circuitry. This can explain the robust and rapid response of ECT in a patient with delirious mania.37,38

Neurotransmitter hypothesis. It has been shown that in patients with delirious mania there is dysregulation of dopamine transport, which leads to dopamine overflow in the synapse. In contrast to a drug effect (ie, cocaine or methamphetamine) that acts by inhibiting dopamine reuptake, dopamine overflow in delirious mania is caused by the loss of dopamine transporter regulation. This results in a dysfunctional dopaminergic state that precipitates an acute state of delirium and agitation.39,40

Serotonin plays a role in mood disorders, including mania and depression.41,42 More specifically, serotonin has been implicated in impulsivity and aggression as shown by reduced levels of CSF 5-hydroxyindoleacetic acid (5-HIAA) and depletion of 5-hydroxytryptophan (5-HTP).43

Continue to: Alterations in gamma-aminobutyric acid (GABA) transmission...

 

 

Alterations in gamma-aminobutyric acid (GABA) transmission are known to occur in delirium and catatonia. In delirium, GABA signaling is increased, which disrupts the circadian rhythm and melatonin release, thus impairing the sleep-wake cycle.44 Deficiencies in acetylcholine and melatonin are seen as well as excess of other neurotransmitters, including norepinephrine and glutamate.45 Conversely, in catatonia, functional imaging studies found decreased GABA-A binding in orbito­frontal, prefrontal, parietal, and motor cortical regions.46 A study analyzing 10 catatonic patients found decreased density of GABA-A receptors in the left sensorimotor cortex compared to psychiatric and healthy controls.47

Other neurotransmitters, such as glutamate, at the N-methyl-D-aspartate receptors (NMDAR) have been hypothesized to be hyperactive, causing downstream dysregulation of GABA functioning.48 However, the exact connection between delirious mania and all these receptors and neurotransmitters remains unknown.

Encephalitis hypothesis. The relationship between delirious mania and autoimmune encephalitis suggests delirious mania has etiologies other than a primary psychiatric illness. In a 2020 retrospective study49 that analyzed 79 patients with anti-NMDAR encephalitis, 25.3% met criteria for delirious mania, and 95% of these patients had catatonic features. Dalmau et al50 found that in many cases, patients tend to respond to ECT; in a cases series of 3 patients, 2 responded to benzodiazepines.

COVID-19 hypothesis. The SARS-CoV-2 virion has been associated with many neuropsychiatric complications, including mood, psychotic, and neurocognitive disorders.51,52 There also have been cases of COVID-19–induced catatonia.53-55 One case of delirious mania in a patient with COVID-19 has been reported.21 The general mechanism has been proposed to be related to the stimulation of the proinflammatory cytokines, such as tumor necrosis factor-alpha and interleukin-6, which the virus produces in large quantities.56 These cytokines have been linked to psychosis and other psychiatric disorders.57 The patient with COVID-19–induced delirious mania had elevated inflammatory markers, including erythrocyte sedimentation rate, C-reactive protein, ferritin, and D-dimer, which supports a proinflammatory state. This patient had a complete resolution of symptoms with ECT.21

Management: Benzodiazepines and ECT

A step-by-step algorithm for managing delirious mania is outlined in the Figure. Regardless of the underlining etiology, management of delirious mania consists of benzodiazepines (lorazepam and diazepam); prompt use of ECT, particularly for patients who do not improve with large doses of lorazepam; or (if applicable) continued treatment of the underlining medical condition, which does not preclude the use of benzodiazepines or ECT. Recent reports27,58 have described details for using ECT for delirious mania, highlighting the use of high-energy dosing, bilateral electrode placement, and frequent sessions.

CP02212026_f1.png

Continue to: Knowing which medications...

 

 

Knowing which medications to avoid is as important as knowing which agents to administer. Be vigilant in avoiding high-potency antipsychotics, as these medications can worsen extrapyramidal symptoms and may precipitate seizures or neuroleptic malignant syndrome (NMS).28 Anticholinergic agents should also be avoided because they worsen confusion. Although lithium is effective in BD, in delirious mania, high doses of lithium and haloperidol may cause severe encephalopathic syndromes, with symptoms that can include lethargy, tremors, cerebellar dysfunction, and worsened confusion; it may also cause widespread and irreversible brain damage.59While positive outcomes have been documented when using a combination of antipsychotics and lithium,8,60 this approach should be considered carefully and tailored to individual cases, taking into account the severity of manic and psychotic symptoms in addition to the level of catatonia.

Due to long periods of hyperactivity, withdrawal, and diaphoresis, patients with delirious mania may be severely dehydrated with metabolic derangements, including elevated CPK due to rhabdomyolysis from prolonged exertion, IM antipsychotics, or rigidity. To prevent acute renal failure, this must be immediately addressed with rapid fluid resuscitation and electrolyte repletion.61

Benzodiazepines. The rapid use of lorazepam should be initiated when delirious mania is suspected. Doses of 6 to 20 mg have been reported to be effective if tolerated.5,20 Typically, high-dose lorazepam will not have the sedative effect that would normally occur in a patient who does not have delirious mania.2 Lorazepam should be titrated until full resolution of symptoms. Doses up to 30 mg have been reported as effective and tolerable.62 In our literature review, 50% of patients (18/36) responded or partially responded to lorazepam. However, only 3 case reports documented a complete remission with lorazepam, and many patients needed ECT for remission of symptoms.

ECT is generally reserved for patients who are not helped by benzodiazepine therapy, which is estimated to be up to 20%.5 ECT is highly effective in delirious mania, with remission rates ranging from 80% to 100%.1 ECT is also effective in acute non­delirious mania (comparable to depression); however, it is only used in a small minority of cases (0.2% to 12%).35 In our review, 58% of cases (21/36) reported using ECT, and in all cases it resulted in complete remission.

A dramatic improvement can be seen even after a single ECT session, though most patients show improvement after 4 sessions or 3 to 7 days.1,2,5 In our review, most patients received 4 to 12 sessions until achieving complete remission.

Continue to: No RCTs have evaluated...

 

 

No RCTs have evaluated ECT electrode placement in patients with delirious mania. However, several RCTs have investigated electrode placement in patients with acute nondelirious mania. Hiremani et al63 found that bitemporal placement had a more rapid response rate than bifrontal placement, but there was no overall difference in response rate. Barekatain et al64 found no difference between these 2 bilateral approaches. Many of the delirious mania cases report using a bilateral placement (including 42% of the ECT cases in our review) due to the emergent need for rapid relief of symptoms, which is especially necessary if the patient is experiencing hemodynamic instability, excessive violence, risk for self-harm, worsening delirium, or resistance to lorazepam.

Prognosis: Often fatal if left untreated

Patients with delirious mania are at high risk to progress to a more severe form of NMS or malignant catatonia. Therefore, high-potency antipsychotics should be avoided; mortality can be elevated from 60% without antipsychotics to 78% with antipsychotics.4 Some researchers estimate 75% to 78% of cases of delirious mania can be fatal if left untreated.3,6

 

Bottom Line

Delirious mania is routinely mistaken for more conventional manic or psychotic disorders. Clinicians need to be able to rapidly recognize the symptoms of this syndrome, which include mania, psychosis, delirium, and possible catatonia, so they can avoid administering toxic agents and instead initiate effective treatments such as benzodiazepines and electroconvulsive therapy.

Related Resources

Drug Brand Names

Diazepam • Valium
Haloperidol • Haldol
Lithium • Eskalith, Lithobid
Lorazepam • Ativan

[embed:render:related:node:249909]

References

1. Fink M. Delirious mania. Bipolar Disord. 1999;1(1):54-60.

2. Karmacharya R, England ML, Ongür D. Delirious mania: clinical features and treatment response. J Affect Disord. 2008;109(3):312-316.

3. Friedman RS, Mufson MJ, Eisenberg TD, et al. Medically and psychiatrically ill: the challenge of delirious mania. Harv Rev Psychiatry. 2003;11(2):91-98.

4. Mann SC, Caroff SN, Bleier HR, et al. Lethal catatonia. Am J Psychiatry. 1986;143(11):1374-1381.

5. Detweiler MB, Mehra A, Rowell T, et al. Delirious mania and malignant catatonia: a report of 3 cases and review. Psychiatr Q. 2009;80(1):23-40.

6. Bell L. On a form of disease resembling some advanced stages of mania and fever. American Journal of Insanity. 1849;6(2):97-127.

7. Carlson GA, Goodwin FK. The stages of mania. A longitudinal analysis of the manic episode. Arch Gen Psychiatry. 1973;28(2):221-228.

8. Bond TC. Recognition of acute delirious mania. Arch Gen Psychiatry. 1980;37(5):553-554.

9. Hutchinson G, David A. Manic pseudo-delirium - two case reports. Behav Neurol. 1997;10(1):21-23.

10. Bush G, Fink M, Petrides G, et al. Catatonia. I. Rating scale and standardized examination. Acta Psychiatr Scand. 1996;93(2):129-136.

11. Bush G, Fink M, Petrides G, et al. Catatonia. II. Treatment with lorazepam and electroconvulsive therapy. Acta Psychiatr Scand. 1996;93(2):137-143.

12. Cordeiro CR, Saraiva R, Côrte-Real B, et al. When the bell rings: clinical features of Bell’s mania. Prim Care Companion CNS Disord. 2020;22(2):19l02511. doi:10.4088/PCC.19l02511

13. Yeo LX, Kuo TC, Hu KC, et al. Lurasidone-induced delirious mania. Am J Ther. 2019;26(6):e786-e787.

14. Jung WY, Lee BD. Quetiapine treatment for delirious mania in a military soldier. Prim Care Companion J Clin Psychiatry. 2010;12(2):PCC.09l00830. doi:10.4088/PCC.09l00830yel

15. Wahid N, Chin G, Turner AH, et al. Clinical response of clozapine as a treatment for delirious mania. Ment Illn. 2017;9(2):7182. doi:10.4081/mi.2017.7182

16. Taylor MA, Fink M. Catatonia in psychiatric classification: a home of its own. Am J Psychiatry. 2003;160(7):1233-1241.

17. Danivas V, Behere RV, Varambally S, et al. Electroconvulsive therapy in the treatment of delirious mania: a report of 2 patients. J ECT. 2010;26(4):278-279.

18. O’Callaghan N, McDonald C, Hallahan B. Delirious mania intractable to treatment. Ir J Psychol Med. 2016;33(2):129-132.

19. Vasudev K, Grunze H. What works for delirious catatonic mania? BMJ Case Rep. 2010;2010:bcr0220102713. doi:10.1136/bcr.02.2010.2713

20. Jacobowski NL, Heckers S, Bobo WV. Delirious mania: detection, diagnosis, and clinical management in the acute setting. J Psychiatr Pract. 2013;19(1):15-28.

21. Reinfeld S, Yacoub A. A case of delirious mania induced by COVID-19 treated with electroconvulsive therapy. J ECT. 2021;37(4):e38-e39.

22. Lee BS, Huang SS, Hsu WY, et al. Clinical features of delirious mania: a series of five cases and a brief literature review. BMC Psychiatry. 2012;12:65. doi:10.1186/1471-244X-12-65

23. Bipeta R, Khan MA. Delirious mania: can we get away with this concept? A case report and review of the literature. Case Rep Psychiatry. 2012;2012:720354. doi:10.1155/2012/720354

24. Nunes AL, Cheniaux E. Delirium and mania with catatonic features in a Brazilian patient: response to ECT. J Neuropsychiatry Clin Neurosci. 2014;26(1):E1-E3.

25. Tegin C, Kalayil G, Lippmann S. Electroconvulsive therapy and delirious catatonic mania. J ECT. 2017;33(4):e33-e34.

26. Melo AL, Serra M. Delirious mania and catatonia. Bipolar Disord. 2020;22(6):647-649.

27. Fink M. Expanding the catatonia tent: recognizing electroconvulsive therapy responsive syndromes. J ECT. 2021;37(2):77-79.

28. Fink M. Electroconvulsive Therapy: A Guide for Professionals and Their Patients. Oxford University Press; 2009.

29. Fink M, Taylor MA. The many varieties of catatonia. Eur Arch Psychiatry Clin Neurosci. 2001;251 Suppl 1:I8-I13.

30. Vivanti A, Harvey K, Ash S, et al. Clinical assessment of dehydration in older people admitted to hospital: what are the strongest indicators? Arch Gerontol Geriatr. 2008;47(3):340-355.

31. Ware MR, Feller DB, Hall KL. Neuroleptic malignant syndrome: diagnosis and management. Prim Care Companion CNS Disord. 2018;20(1):17r02185. doi:10.4088/PCC.17r0218

32. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189-198.

33. Taylor MA, Abrams R. The phenomenology of mania. A new look at some old patients. Arch Gen Psychiatry. 1973;29(4):520-522.

34. Klerman GL. The spectrum of mania. Compr Psychiatry. 1981;22(1):11-20.

35. Elias A, Thomas N, Sackeim HA. Electroconvulsive therapy in mania: a review of 80 years of clinical experience. Am J Psychiatry. 2021;178(3):229-239.

36. Thom RP, Levy-Carrick NC, Bui M, et al. Delirium. Am J Psychiatry. 2019;176(10):785-793.

37. Charlton BG, Kavanau JL. Delirium and psychotic symptoms--an integrative model. Med Hypotheses. 2002;58(1):24-27.

38. Kramp P, Bolwig TG. Electroconvulsive therapy in acute delirious states. Compr Psychiatry. 1981;22(4):368-371.

39. Mash DC. Excited delirium and sudden death: a syndromal disorder at the extreme end of the neuropsychiatric continuum. Front Physiol. 2016;7:435.

40. Strawn JR, Keck PE Jr, Caroff SN. Neuroleptic malignant syndrome. Am J Psychiatry. 2007;164(6):870-876.

41. Charney DS. Monoamine dysfunction and the pathophysiology and treatment of depression. J Clin Psychiatry. 1998;59 Suppl 14:11-14.

42. Shiah IS, Yatham LN. Serotonin in mania and in the mechanism of action of mood stabilizers: a review of clinical studies. Bipolar Disord. 2000;2(2):77-92.

43. Dalley JW, Roiser JP. Dopamine, serotonin and impulsivity. Neuroscience. 2012;215:42-58.

44. Maldonado JR. Pathoetiological model of delirium: a comprehensive understanding of the neurobiology of delirium and an evidence-based approach to prevention and treatment. Crit Care Clin. 2008;24(4):789-856, ix.

45. Maldonado JR. Neuropathogenesis of delirium: review of current etiologic theories and common pathways. Am J Geriatr Psychiatry. 2013;21(12):1190-1222.

46. Rasmussen SA, Mazurek MF, Rosebush PI. Catatonia: our current understanding of its diagnosis, treatment and pathophysiology. World J Psychiatry. 2016;6(4):391-398.

47. Northoff G, Steinke R, Czcervenka C, et al. Decreased density of GABA-A receptors in the left sensorimotor cortex in akinetic catatonia: investigation of in vivo benzodiazepine receptor binding. J Neurol Neurosurg Psychiatry. 1999;67(4):445-450.

48. Daniels J. Catatonia: clinical aspects and neurobiological correlates. J Neuropsychiatry Clin Neurosci. 2009;21(4):371-380.

49. Restrepo-Martínez M, Chacón-González J, Bayliss L, et al. Delirious mania as a neuropsychiatric presentation in patients with anti-N-methyl-D-aspartate receptor encephalitis. Psychosomatics. 2020;61(1):64-69.

50. Dalmau J, Armangué T, Planagumà J, et al. An update on anti-NMDA receptor encephalitis for neurologists and psychiatrists: mechanisms and models. Lancet Neurol. 2019;18(11):1045-1057.

51. Steardo L Jr, Steardo L, Verkhratsky A. Psychiatric face of COVID-19. Transl Psychiatry. 2020;10(1):261.

52. Iqbal Y, Al Abdulla MA, Albrahim S, et al. Psychiatric presentation of patients with acute SARS-CoV-2 infection: a retrospective review of 50 consecutive patients seen by a consultation-liaison psychiatry team. BJPsych Open. 2020;6(5):e109.

53. Gouse BM, Spears WE, Nieves Archibald A, et al. Catatonia in a hospitalized patient with COVID-19 and proposed immune-mediated mechanism. Brain Behav Immun. 2020;89:529-530.

54. Caan MP, Lim CT, Howard M. A case of catatonia in a man with COVID-19. Psychosomatics. 2020;61(5):556-560.

55. Zain SM, Muthukanagaraj P, Rahman N. Excited catatonia - a delayed neuropsychiatric complication of COVID-19 infection. Cureus. 2021;13(3):e13891.

56. Chowdhury MA, Hossain N, Kashem MA, et al. Immune response in COVID-19: a review. J Infect Public Health. 2020;13(11):1619-1629.

57. Radhakrishnan R, Kaser M, Guloksuz S. The link between the immune system, environment, and psychosis. Schizophr Bull. 2017;43(4):693-697.

58. Fink M, Kellner CH, McCall WV. Optimizing ECT technique in treating catatonia. J ECT. 2016;32(3):149-150.

59. Cohen WJ, Cohen NH. Lithium carbonate, haloperidol, and irreversible brain damage. JAMA. 1974;230(9):1283-1287.

60. Davis MJ, de Nesnera A, Folks DG. Confused and nearly naked after going on spending sprees. Current Psychiatry. 2014;13(7):56-62.

61. Stanley M, Chippa V, Aeddula NR, et al. Rhabdomyolysis. StatPearls Publishing; 2021.

62. Fink M, Taylor MA. The catatonia syndrome: forgotten but not gone. Arch Gen Psychiatry. 2009;66(11):1173-1177.

63. Hiremani RM, Thirthalli J, Tharayil BS, et al. Double-blind randomized controlled study comparing short-term efficacy of bifrontal and bitemporal electroconvulsive therapy in acute mania. Bipolar Disord. 2008;10(6):701-707.

64. Barekatain M, Jahangard L, Haghighi M, et al. Bifrontal versus bitemporal electroconvulsive therapy in severe manic patients. J ECT. 2008;24(3):199-202.

References

1. Fink M. Delirious mania. Bipolar Disord. 1999;1(1):54-60.

2. Karmacharya R, England ML, Ongür D. Delirious mania: clinical features and treatment response. J Affect Disord. 2008;109(3):312-316.

3. Friedman RS, Mufson MJ, Eisenberg TD, et al. Medically and psychiatrically ill: the challenge of delirious mania. Harv Rev Psychiatry. 2003;11(2):91-98.

4. Mann SC, Caroff SN, Bleier HR, et al. Lethal catatonia. Am J Psychiatry. 1986;143(11):1374-1381.

5. Detweiler MB, Mehra A, Rowell T, et al. Delirious mania and malignant catatonia: a report of 3 cases and review. Psychiatr Q. 2009;80(1):23-40.

6. Bell L. On a form of disease resembling some advanced stages of mania and fever. American Journal of Insanity. 1849;6(2):97-127.

7. Carlson GA, Goodwin FK. The stages of mania. A longitudinal analysis of the manic episode. Arch Gen Psychiatry. 1973;28(2):221-228.

8. Bond TC. Recognition of acute delirious mania. Arch Gen Psychiatry. 1980;37(5):553-554.

9. Hutchinson G, David A. Manic pseudo-delirium - two case reports. Behav Neurol. 1997;10(1):21-23.

10. Bush G, Fink M, Petrides G, et al. Catatonia. I. Rating scale and standardized examination. Acta Psychiatr Scand. 1996;93(2):129-136.

11. Bush G, Fink M, Petrides G, et al. Catatonia. II. Treatment with lorazepam and electroconvulsive therapy. Acta Psychiatr Scand. 1996;93(2):137-143.

12. Cordeiro CR, Saraiva R, Côrte-Real B, et al. When the bell rings: clinical features of Bell’s mania. Prim Care Companion CNS Disord. 2020;22(2):19l02511. doi:10.4088/PCC.19l02511

13. Yeo LX, Kuo TC, Hu KC, et al. Lurasidone-induced delirious mania. Am J Ther. 2019;26(6):e786-e787.

14. Jung WY, Lee BD. Quetiapine treatment for delirious mania in a military soldier. Prim Care Companion J Clin Psychiatry. 2010;12(2):PCC.09l00830. doi:10.4088/PCC.09l00830yel

15. Wahid N, Chin G, Turner AH, et al. Clinical response of clozapine as a treatment for delirious mania. Ment Illn. 2017;9(2):7182. doi:10.4081/mi.2017.7182

16. Taylor MA, Fink M. Catatonia in psychiatric classification: a home of its own. Am J Psychiatry. 2003;160(7):1233-1241.

17. Danivas V, Behere RV, Varambally S, et al. Electroconvulsive therapy in the treatment of delirious mania: a report of 2 patients. J ECT. 2010;26(4):278-279.

18. O’Callaghan N, McDonald C, Hallahan B. Delirious mania intractable to treatment. Ir J Psychol Med. 2016;33(2):129-132.

19. Vasudev K, Grunze H. What works for delirious catatonic mania? BMJ Case Rep. 2010;2010:bcr0220102713. doi:10.1136/bcr.02.2010.2713

20. Jacobowski NL, Heckers S, Bobo WV. Delirious mania: detection, diagnosis, and clinical management in the acute setting. J Psychiatr Pract. 2013;19(1):15-28.

21. Reinfeld S, Yacoub A. A case of delirious mania induced by COVID-19 treated with electroconvulsive therapy. J ECT. 2021;37(4):e38-e39.

22. Lee BS, Huang SS, Hsu WY, et al. Clinical features of delirious mania: a series of five cases and a brief literature review. BMC Psychiatry. 2012;12:65. doi:10.1186/1471-244X-12-65

23. Bipeta R, Khan MA. Delirious mania: can we get away with this concept? A case report and review of the literature. Case Rep Psychiatry. 2012;2012:720354. doi:10.1155/2012/720354

24. Nunes AL, Cheniaux E. Delirium and mania with catatonic features in a Brazilian patient: response to ECT. J Neuropsychiatry Clin Neurosci. 2014;26(1):E1-E3.

25. Tegin C, Kalayil G, Lippmann S. Electroconvulsive therapy and delirious catatonic mania. J ECT. 2017;33(4):e33-e34.

26. Melo AL, Serra M. Delirious mania and catatonia. Bipolar Disord. 2020;22(6):647-649.

27. Fink M. Expanding the catatonia tent: recognizing electroconvulsive therapy responsive syndromes. J ECT. 2021;37(2):77-79.

28. Fink M. Electroconvulsive Therapy: A Guide for Professionals and Their Patients. Oxford University Press; 2009.

29. Fink M, Taylor MA. The many varieties of catatonia. Eur Arch Psychiatry Clin Neurosci. 2001;251 Suppl 1:I8-I13.

30. Vivanti A, Harvey K, Ash S, et al. Clinical assessment of dehydration in older people admitted to hospital: what are the strongest indicators? Arch Gerontol Geriatr. 2008;47(3):340-355.

31. Ware MR, Feller DB, Hall KL. Neuroleptic malignant syndrome: diagnosis and management. Prim Care Companion CNS Disord. 2018;20(1):17r02185. doi:10.4088/PCC.17r0218

32. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189-198.

33. Taylor MA, Abrams R. The phenomenology of mania. A new look at some old patients. Arch Gen Psychiatry. 1973;29(4):520-522.

34. Klerman GL. The spectrum of mania. Compr Psychiatry. 1981;22(1):11-20.

35. Elias A, Thomas N, Sackeim HA. Electroconvulsive therapy in mania: a review of 80 years of clinical experience. Am J Psychiatry. 2021;178(3):229-239.

36. Thom RP, Levy-Carrick NC, Bui M, et al. Delirium. Am J Psychiatry. 2019;176(10):785-793.

37. Charlton BG, Kavanau JL. Delirium and psychotic symptoms--an integrative model. Med Hypotheses. 2002;58(1):24-27.

38. Kramp P, Bolwig TG. Electroconvulsive therapy in acute delirious states. Compr Psychiatry. 1981;22(4):368-371.

39. Mash DC. Excited delirium and sudden death: a syndromal disorder at the extreme end of the neuropsychiatric continuum. Front Physiol. 2016;7:435.

40. Strawn JR, Keck PE Jr, Caroff SN. Neuroleptic malignant syndrome. Am J Psychiatry. 2007;164(6):870-876.

41. Charney DS. Monoamine dysfunction and the pathophysiology and treatment of depression. J Clin Psychiatry. 1998;59 Suppl 14:11-14.

42. Shiah IS, Yatham LN. Serotonin in mania and in the mechanism of action of mood stabilizers: a review of clinical studies. Bipolar Disord. 2000;2(2):77-92.

43. Dalley JW, Roiser JP. Dopamine, serotonin and impulsivity. Neuroscience. 2012;215:42-58.

44. Maldonado JR. Pathoetiological model of delirium: a comprehensive understanding of the neurobiology of delirium and an evidence-based approach to prevention and treatment. Crit Care Clin. 2008;24(4):789-856, ix.

45. Maldonado JR. Neuropathogenesis of delirium: review of current etiologic theories and common pathways. Am J Geriatr Psychiatry. 2013;21(12):1190-1222.

46. Rasmussen SA, Mazurek MF, Rosebush PI. Catatonia: our current understanding of its diagnosis, treatment and pathophysiology. World J Psychiatry. 2016;6(4):391-398.

47. Northoff G, Steinke R, Czcervenka C, et al. Decreased density of GABA-A receptors in the left sensorimotor cortex in akinetic catatonia: investigation of in vivo benzodiazepine receptor binding. J Neurol Neurosurg Psychiatry. 1999;67(4):445-450.

48. Daniels J. Catatonia: clinical aspects and neurobiological correlates. J Neuropsychiatry Clin Neurosci. 2009;21(4):371-380.

49. Restrepo-Martínez M, Chacón-González J, Bayliss L, et al. Delirious mania as a neuropsychiatric presentation in patients with anti-N-methyl-D-aspartate receptor encephalitis. Psychosomatics. 2020;61(1):64-69.

50. Dalmau J, Armangué T, Planagumà J, et al. An update on anti-NMDA receptor encephalitis for neurologists and psychiatrists: mechanisms and models. Lancet Neurol. 2019;18(11):1045-1057.

51. Steardo L Jr, Steardo L, Verkhratsky A. Psychiatric face of COVID-19. Transl Psychiatry. 2020;10(1):261.

52. Iqbal Y, Al Abdulla MA, Albrahim S, et al. Psychiatric presentation of patients with acute SARS-CoV-2 infection: a retrospective review of 50 consecutive patients seen by a consultation-liaison psychiatry team. BJPsych Open. 2020;6(5):e109.

53. Gouse BM, Spears WE, Nieves Archibald A, et al. Catatonia in a hospitalized patient with COVID-19 and proposed immune-mediated mechanism. Brain Behav Immun. 2020;89:529-530.

54. Caan MP, Lim CT, Howard M. A case of catatonia in a man with COVID-19. Psychosomatics. 2020;61(5):556-560.

55. Zain SM, Muthukanagaraj P, Rahman N. Excited catatonia - a delayed neuropsychiatric complication of COVID-19 infection. Cureus. 2021;13(3):e13891.

56. Chowdhury MA, Hossain N, Kashem MA, et al. Immune response in COVID-19: a review. J Infect Public Health. 2020;13(11):1619-1629.

57. Radhakrishnan R, Kaser M, Guloksuz S. The link between the immune system, environment, and psychosis. Schizophr Bull. 2017;43(4):693-697.

58. Fink M, Kellner CH, McCall WV. Optimizing ECT technique in treating catatonia. J ECT. 2016;32(3):149-150.

59. Cohen WJ, Cohen NH. Lithium carbonate, haloperidol, and irreversible brain damage. JAMA. 1974;230(9):1283-1287.

60. Davis MJ, de Nesnera A, Folks DG. Confused and nearly naked after going on spending sprees. Current Psychiatry. 2014;13(7):56-62.

61. Stanley M, Chippa V, Aeddula NR, et al. Rhabdomyolysis. StatPearls Publishing; 2021.

62. Fink M, Taylor MA. The catatonia syndrome: forgotten but not gone. Arch Gen Psychiatry. 2009;66(11):1173-1177.

63. Hiremani RM, Thirthalli J, Tharayil BS, et al. Double-blind randomized controlled study comparing short-term efficacy of bifrontal and bitemporal electroconvulsive therapy in acute mania. Bipolar Disord. 2008;10(6):701-707.

64. Barekatain M, Jahangard L, Haghighi M, et al. Bifrontal versus bitemporal electroconvulsive therapy in severe manic patients. J ECT. 2008;24(3):199-202.

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
26-34,54
Page Number
26-34,54
Publications
Publications
Topics
Article Type
Display Headline
Delirious mania: Presentation, pathogenesis, and management
Display Headline
Delirious mania: Presentation, pathogenesis, and management
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1223 Reinfeld</fileName> <TBEID>0C02E901.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02E901</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Delirious mania: Presentation, p</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231129T075706</firstPublished> <LastPublished>20231129T075706</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231129T075705</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Samuel Reinfeld, DO, Adeeb Yacoub, MD</byline> <bylineText/> <bylineFull>Samuel Reinfeld, DO, Adeeb Yacoub, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>26-34,54</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Delirious mania is a syndrome characterized by the acute onset of severe hyperactivity, psychosis, catatonia, and intermittent confusion. While there have been </metaDescription> <articlePDF>299295</articlePDF> <teaserImage/> <title>Delirious mania: Presentation, pathogenesis, and management</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords> <keyword>bipolar disorder</keyword> <keyword> schizophrenia</keyword> <keyword> delirious mania</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">26935</term> </sections> <topics> <term canonical="true">190</term> <term>293</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002644.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Delirious mania: Presentation, pathogenesis, and management</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Rapid recognition is critical to avoid initiating inappropriate treatment</p> <p>Delirious mania is a syndrome characterized by the acute onset of severe hyperactivity, psychosis, catatonia, and intermittent confusion. While there have been growing reports of this phenomenon over the last 2 decades, it remains poorly recognized and understood.<sup>1,2</sup> There is no widely accepted nosology for delirious mania and the condition is absent from DSM-5, which magnifies the difficulties in making a timely diagnosis and initiating appropriate treatment. Delayed diagnosis and treatment may result in a detrimental outcome.<sup>2,3</sup> Delirious mania has also been labeled as lethal catatonia, specific febrile delirium, hyperactive or exhaustive mania, and Bell’s mania.<sup>2,4,5</sup> The characterization and diagnosis of this condition have a long and inconsistent history (<span class="bitalic">Box</span>,<sup>1,6-11</sup> <span class="bitalic">page 28</span>).</p> <p>Delirious mania continues to be met with incertitude in clinical practice, and numerous inconsistencies have been reported in the literature. For example, some cases that have been reported as delirious mania had more evidence of primary delirium due to another medical condition or primary mania.<sup>12,13</sup> Other cases have demonstrated swift improvement of symptoms after monotherapy with antipsychotics without a trial of benzodiazepines or electroconvulsive therapy (ECT); the exclusion of a sudden onset questions the validity of the diagnosis and promotes the use of less efficacious treatments.<sup>14,15</sup> Other reports have confirmed that the diagnosis is missed when certain symptoms are more predominant, such as a thought disorder (acute schizophrenia), grandiosity and delusional ideation (bipolar disorder [BD]), and less commonly assessed catatonic signs (ambitendency, automatic obedience). These symptoms are mistakenly attributed to the respective disease.<sup>1,16</sup> This especially holds true when delirious mania is initially diagnosed as a primary psychosis, which leads to the administration of antipsychotics.<sup>17</sup> Other cases have reported that delirious mania was resistant to treatment, but ECT was never pursued.<sup>18</sup>In this review, we provide a more comprehensive perspective of the clinical presentation, pathogenesis, and management of delirious mania. We searched PubMed and Google Scholar using the keywords “delirious mania,” “delirious mania AND catatonia,” or “manic delirium.” Most articles we found were case reports, case series, or retrospective chart reviews. There were no systematic reviews, meta analyses, or randomized control trials (RCTs). The 12 articles included in this review consist of 7 individual case reports, 4 case series, and 1 retrospective chart review that describe a total of 36 cases (<b><i>Table</i></b>,<sup>1,2,5,17,19-26</sup> <b><i>page 30</i></b>).</p> <h3>Clinical presentation: What to look for</h3> <p>Patients with delirious mania typically develop symptoms extremely rapidly. In virtually all published literature, symptoms were reported to emerge within hours to days and consisted of severe forms of mania, psychosis, and delirium; 100% of the cases in our review had these symptoms. Commonly reported symptoms were:</p> <ul class="body"> <li>intense excitement</li> <li>emotional lability</li> <li>grandiose delusions</li> <li>profound insomnia</li> <li>pressured and rapid speech</li> <li>auditory and visual hallucinations</li> <li>hypersexuality</li> <li>thought disorganization.</li> </ul> <p>Exquisite paranoia can also result in violent aggression (and may require the use of physical restraints). Patients may confine themselves to very small spaces (such as a closet) in response to the intense paranoia. Impairments in various neurocognitive domains—including inability to focus; disorientation; language and visuospatial disturbances; difficulty with shifting and sustaining attention; and short-term memory impairments—have been reported. Patients often cannot recall the events during the episode.<sup>1,2,5,27,28</sup><br/><br/>Catatonia has been closely associated with delirious mania.<sup>29</sup> Features of excited catatonia—such as excessive motor activity, negativism, grimacing, posturing, echolalia, echopraxia, stereotypy, automatic obedience, verbigeration, combativeness, impulsivity, and rigidity—typically accompany delirious mania.<sup>1,5,10,19,27</sup><br/><br/>In addition to these symptoms, patients may engage in specific behaviors. They may exhibit inappropriate toileting such as smearing feces on walls or in bags, fecal or urinary incontinence, disrobing or running naked in public places, or pouring liquid on the floor or on one’s head.<sup>1,2</sup>Of the 36 cases reported in the literature we reviewed, 20 (55%) were female. Most patients had an underlining psychiatric condition, including BD (72%), major depressive disorder (8%), and schizophrenia (2%). Three patients had no psychiatric history.</p> <h3>Physical examination</h3> <p>On initial presentation, a patient with delirious mania may be dehydrated, with dry mucous membranes, pale conjunctiva, tongue dryness, and poor skin turgor.<sup>28,30</sup> Due to excessive motor activity, diaphoresis with tachycardia, fluctuating blood pressure, and fever may be present.<sup>31</sup></p> <p>Certain basic cognitive tasks should be assessed to determine the patient’s orientation to place, date, and time. Assess if the patient can recall recent events, names of objects, or perform serial 7s; clock drawing capabilities also should be ascertained.<sup>1,2,5</sup> A Mini-Mental State Examination is useful.<sup>32</sup><br/><br/>The Bush-Francis Catatonia Rating Scale should be used to elicit features of catatonia, such as waxy flexibility, negativism, gegenhalten, mitgehen, catalepsy, ambitendency, automatic obedience, and grasp reflex.<sup>10</sup></p> <h3>Laboratory findings are nonspecific</h3> <p>Although no specific laboratory findings are associated with delirious mania, bloodwork and imaging are routinely investigated, especially if delirium characteristics are most striking. A complete blood count, chemistries, hepatic panel, thyroid functioning, blood and/or urine cultures, creatinine phosphokinase (CPK), and urinalysis can be ordered. Head imaging such as MRI and CT to rule out intracranial pathology are typically performed.<sup>19</sup> However, the diagnosis of delirious mania is based on the presence of the phenotypic features, by verification of catatonia, and by the responsiveness to the treatment delivered.<sup>29</sup></p> <h3>Pathogenesis: Several hypotheses</h3> <p>The pathogenesis of delirious mania is not well understood. There are several postulations but no salient theory. Most patients with delirious mania have an underlying systemic medical or psychiatric condition. </p> <p><span class="bbody">Mood disorders.</span> Patients with BD or schizoaffective disorder are especially susceptible to delirious mania. The percentage of manic patients who present with delirious mania varies by study. One study suggested approximately 19% have features of the phenomenon,<sup>33</sup> while others estimated 15% to 25%.<sup>34</sup> Elias et al<sup>35</sup> calculated that 15% of patients with mania succumb to manic exhaustion; from this it can be reasonably concluded that these were cases of misdiagnosed delirious mania.<span class="bbody">Delirium hypothesis.</span> Patients with delirious mania typically have features of delirium, including fluctuation of consciousness, disorientation, and/or poor sleep-wake cycle.<sup>36</sup> During rapid eye movement (REM) and non-REM sleep, memory circuits are fortified. When there is a substantial loss of REM and non-REM sleep, these circuits become faulty, even after 1 night. Pathological brain waves on EEG reflect the inability to reinforce the memory circuits. Patients with these waves may develop hallucinations, bizarre delusions, and altered sensorium. ECT reduces the pathological slow wave morphologies, thus restoring the synaptic maintenance and correcting the incompetent circuitry. This can explain the robust and rapid response of ECT in a patient with delirious mania.<sup>37,38</sup><span class="bbody">Neurotransmitter hypothesis.</span><b> </b>It has been shown that in patients with delirious mania there is dysregulation of dopamine transport, which leads to dopamine overflow in the synapse. In contrast to a drug effect (ie, cocaine or methamphetamine) that acts by inhibiting dopamine reuptake, dopamine overflow in delirious mania is caused by the loss of dopamine transporter regulation. This results in a dysfunctional dopaminergic state that precipitates an acute state of delirium and agitation.<sup>39,40</sup>Serotonin plays a role in mood disorders, including mania and depression.<sup>41,42</sup> More specifically, serotonin has been implicated in impulsivity and aggression as shown by reduced levels of CSF 5-hydroxyindoleacetic acid (5-HIAA) and depletion of 5-hydroxytryptophan (5-HTP).<sup>43</sup>Alterations in gamma-aminobutyric acid (GABA) transmission are known to occur in delirium and catatonia. In delirium, GABA signaling is increased, which disrupts the circadian rhythm and melatonin release, thus impairing the sleep-wake cycle.<sup>44</sup> Deficiencies in acetylcholine and melatonin are seen as well as excess of other neurotransmitters, including norepinephrine and glutamate.<sup>45</sup> Conversely, in catatonia, functional imaging studies found decreased GABA-A binding in orbito­frontal, prefrontal, parietal, and motor cortical regions.<sup>46</sup> A study analyzing 10 catatonic patients found decreased density of GABA-A receptors in the left sensorimotor cortex compared to psychiatric and healthy controls.<sup>47</sup>Other neurotransmitters, such as glutamate, at the <i>N</i>-methyl-<scaps>d</scaps>-aspartate receptors (NMDAR) have been hypothesized to be hyperactive, causing downstream dysregulation of GABA functioning.<sup>48</sup> However, the exact connection between delirious mania and all these receptors and neurotransmitters remains unknown.<span class="bbody">Encephalitis hypothesis.</span> The relationship between delirious mania and autoimmune encephalitis suggests delirious mania has etiologies other than a primary psychiatric illness. In a 2020 retrospective study<sup>49</sup> that analyzed 79 patients with anti-NMDAR encephalitis, 25.3% met criteria for delirious mania, and 95% of these patients had catatonic features. Dalmau et al<sup>50</sup> found that in many cases, patients tend to respond to ECT; in a cases series of 3 patients, 2 responded to benzodiazepines.<br/><br/><span class="bbody">COVID-19 hypothesis.</span> The SARS-CoV-2 virion has been associated with many neuropsychiatric complications, including mood, psychotic, and neurocognitive disorders.<sup>51,52</sup> There also have been cases of COVID-19–induced catatonia.<sup>53-55</sup> One case of delirious mania in a patient with COVID-19 has been reported.<sup>21</sup> The general mechanism has been proposed to be related to the stimulation of the proinflammatory cytokines, such as tumor necrosis factor-alpha and interleukin-6, which the virus produces in large quantities.<sup>56</sup> These cytokines have been linked to psychosis and other psychiatric disorders.<sup>57</sup> The patient with COVID-19–induced delirious mania had elevated inflammatory markers, including erythrocyte sedimentation rate, C-reactive protein, ferritin, and D-dimer, which supports a proinflammatory state. This patient had a complete resolution of symptoms with ECT.<sup>21</sup></p> <h3>Management: Benzodiazepines and ECT</h3> <p>A step-by-step algorithm for managing delirious mania is outlined in the <b><i>Figure </i></b>(<span class="bitalic">page 32</span>). Regardless of the underlining etiology, management of delirious mania consists of benzodiazepines (lorazepam and diazepam); prompt use of ECT, particularly for patients who do not improve with large doses of lorazepam; or (if applicable) continued treatment of the underlining medical condition, which does not preclude the use of benzodiazepines or ECT. Recent reports<sup>27,58</sup> have described details for using ECT for delirious mania, highlighting the use of high-energy dosing, bilateral electrode placement, and frequent sessions.</p> <p>Knowing which medications to avoid is as important as knowing which agents to administer. Be vigilant in avoiding high-potency antipsychotics, as these medications can worsen extrapyramidal symptoms and may precipitate seizures or neuroleptic malignant syndrome (NMS).<sup>28</sup> Anticholinergic agents should also be avoided because they worsen confusion. Although lithium is effective in BD, in delirious mania, high doses of lithium and haloperidol may cause severe encephalopathic syndromes, with symptoms that can include lethargy, tremors, cerebellar dysfunction, and worsened confusion; it may also cause widespread and irreversible brain damage.<sup>59</sup> <hl name="1"/>While positive outcomes have been documented when using a combination of antipsychotics and lithium,<sup>8,60</sup> this approach should be considered carefully and tailored to individual cases, taking into account the severity of manic and psychotic symptoms in addition to the level of catatonia.Due to long periods of hyperactivity, withdrawal, and diaphoresis, patients with delirious mania may be severely dehydrated with metabolic derangements, including elevated CPK due to rhabdomyolysis from prolonged exertion, IM antipsychotics, or rigidity. To prevent acute renal failure, this must be immediately addressed with rapid fluid resuscitation and electrolyte repletion.<sup>61</sup><span class="bbody">Benzodiazepines.</span><b> </b>The rapid use of lorazepam should be initiated when delirious mania is suspected. Doses of 6 to 20 mg have been reported to be effective if tolerated.<sup>5,20</sup> Typically, high-dose lorazepam will not have the sedative effect that would normally occur in a patient who does not have delirious mania.<sup>2</sup> Lorazepam should be titrated until full resolution of symptoms. Doses up to 30 mg have been reported as effective and tolerable.<sup>62</sup> In our literature review, 50% of patients (18/36) responded or partially responded to lorazepam. However, only 3 case reports documented a complete remission with lorazepam, and many patients needed ECT for remission of symptoms.<br/><br/><span class="bbody">ECT </span>is generally reserved for patients who are not helped by benzodiazepine therapy, which is estimated to be up to 20%.<sup>5</sup> ECT is highly effective in delirious mania, with remission rates ranging from 80% to 100%.<sup>1</sup> ECT is also effective in acute non­delirious mania (comparable to depression); however, it is only used in a small minority of cases (0.2% to 12%).<sup>35</sup> In our review, 58% of cases (21/36) reported using ECT, and in all cases it resulted in complete remission. <br/><br/>A dramatic improvement can be seen even after a single ECT session, though most patients show improvement after 4 sessions or 3 to 7 days.<sup>1,2,5</sup> In our review, most patients received 4 to 12 sessions until achieving complete remission.<br/><br/>No RCTs have evaluated ECT electrode placement in patients with delirious mania. However, several RCTs have investigated electrode placement in patients with acute nondelirious mania. Hiremani et al<sup>63</sup> found that bitemporal placement had a more rapid response rate than bifrontal placement, but there was no overall difference in response rate. Barekatain et al<sup>64</sup> found no difference between these 2 bilateral approaches. Many of the delirious mania cases report using a bilateral placement (including 42% of the ECT cases in our review) due to the emergent need for rapid relief of symptoms, which is especially necessary if the patient is experiencing hemodynamic instability, excessive violence, risk for self-harm, worsening delirium, or resistance to lorazepam.</p> <h3>Prognosis: Often fatal if left untreated</h3> <p>Patients with delirious mania are at high risk to progress to a more severe form of NMS or malignant catatonia. Therefore, high-potency antipsychotics should be avoided; mortality can be elevated from 60% without antipsychotics to 78% with antipsychotics.<sup>4</sup> Some researchers estimate 75% to 78% of cases of delirious mania can be fatal if left untreated.<sup>3,6</sup></p> <p> <strong>References</strong> </p> <p class="reference"> 1. Fink M. Delirious mania. Bipolar Disord. 1999;1(1):54-60.<br/><br/> 2. Karmacharya R, England ML, Ongür D. Delirious mania: clinical features and treatment response. J Affect Disord. 2008;109(3):312-316.<br/><br/> 3. Friedman RS, Mufson MJ, Eisenberg TD, et al. Medically and psychiatrically ill: the challenge of delirious mania. Harv Rev Psychiatry. 2003;11(2):91-98.<br/><br/> 4. Mann SC, Caroff SN, Bleier HR, et al. Lethal catatonia. Am J Psychiatry. 1986;143(11):1374-1381.<br/><br/> 5. Detweiler MB, Mehra A, Rowell T, et al. Delirious mania and malignant catatonia: a report of 3 cases and review. Psychiatr Q. 2009;80(1):23-40.<br/><br/> 6. Bell L. On a form of disease resembling some advanced stages of mania and fever. American Journal of Insanity. 1849;6(2):97-127.<br/><br/> 7. Carlson GA, Goodwin FK. The stages of mania. A longitudinal analysis of the manic episode. Arch Gen Psychiatry. 1973;28(2):221-228.<br/><br/> 8. Bond TC. Recognition of acute delirious mania. Arch Gen Psychiatry. 1980;37(5):553-554.<br/><br/> 9. Hutchinson G, David A. Manic pseudo-delirium - two case reports. Behav Neurol. 1997;10(1):21-23.<br/><br/> 10. Bush G, Fink M, Petrides G, et al. Catatonia. I. Rating scale and standardized examination. Acta Psychiatr Scand. 1996;93(2):129-136.<br/><br/> 11. Bush G, Fink M, Petrides G, et al. Catatonia. II. Treatment with lorazepam and electroconvulsive therapy. Acta Psychiatr Scand. 1996;93(2):137-143.<br/><br/> 12. Cordeiro CR, Saraiva R, Côrte-Real B, et al. When the bell rings: clinical features of Bell’s mania. Prim Care Companion CNS Disord. 2020;22(2):19l02511. doi:10.4088/PCC.19l02511<br/><br/> 13. Yeo LX, Kuo TC, Hu KC, et al. Lurasidone-induced delirious mania. Am J Ther. 2019;26(6):e786-e787.<br/><br/> 14. Jung WY, Lee BD. Quetiapine treatment for delirious mania in a military soldier. Prim Care Companion J Clin Psychiatry. 2010;12(2):PCC.09l00830. doi:10.4088/PCC.09l00830yel<br/><br/> 15. Wahid N, Chin G, Turner AH, et al. Clinical response of clozapine as a treatment for delirious mania. Ment Illn. 2017;9(2):7182. doi:10.4081/mi.2017.7182<br/><br/> 16. Taylor MA, Fink M. Catatonia in psychiatric classification: a home of its own. Am J Psychiatry. 2003;160(7):1233-1241.<br/><br/> 17. Danivas V, Behere RV, Varambally S, et al. Electroconvulsive therapy in the treatment of delirious mania: a report of 2 patients. J ECT. 2010;26(4):278-279.<br/><br/> 18. O’Callaghan N, McDonald C, Hallahan B. Delirious mania intractable to treatment. Ir J Psychol Med. 2016;33(2):129-132.<br/><br/> 19. Vasudev K, Grunze H. What works for delirious catatonic mania? BMJ Case Rep. 2010;2010:bcr0220102713. doi:10.1136/bcr.02.2010.2713<br/><br/> 20. Jacobowski NL, Heckers S, Bobo WV. Delirious mania: detection, diagnosis, and clinical management in the acute setting. J Psychiatr Pract. 2013;19(1):15-28.<br/><br/> 21. Reinfeld S, Yacoub A. A case of delirious mania induced by COVID-19 treated with electroconvulsive therapy. J ECT. 2021;37(4):e38-e39.<br/><br/> 22. Lee BS, Huang SS, Hsu WY, et al. Clinical features of delirious mania: a series of five cases and a brief literature review. BMC Psychiatry. 2012;12:65. doi:10.1186/1471-244X-12-65<br/><br/> 23. Bipeta R, Khan MA. Delirious mania: can we get away with this concept? A case report and review of the literature. Case Rep Psychiatry. 2012;2012:720354. doi:10.1155/2012/720354 <br/><br/> 24. Nunes AL, Cheniaux E. Delirium and mania with catatonic features in a Brazilian patient: response to ECT. J Neuropsychiatry Clin Neurosci. 2014;26(1):E1-E3.<br/><br/> 25. Tegin C, Kalayil G, Lippmann S. Electroconvulsive therapy and delirious catatonic mania. J ECT. 2017;33(4):e33-e34.<br/><br/> 26. Melo AL, Serra M. Delirious mania and catatonia. Bipolar Disord. 2020;22(6):647-649.<br/><br/> 27. Fink M. Expanding the catatonia tent: recognizing electroconvulsive therapy responsive syndromes. J ECT. 2021;37(2):77-79.<br/><br/> 28. Fink M. <span class="iref">Electroconvulsive Therapy: A Guide for Professionals and Their Patients</span>. Oxford University Press; 2009.<br/><br/> 29. Fink M, Taylor MA. The many varieties of catatonia. Eur Arch Psychiatry Clin Neurosci. 2001;251 Suppl 1:I8-I13.<br/><br/> 30. Vivanti A, Harvey K, Ash S, et al. Clinical assessment of dehydration in older people admitted to hospital: what are the strongest indicators? Arch Gerontol Geriatr. 2008;47(3):340-355.<br/><br/> 31. Ware MR, Feller DB, Hall KL. Neuroleptic malignant syndrome: diagnosis and management. Prim Care Companion CNS Disord. 2018;20(1):17r02185. doi:10.4088/PCC.17r0218<br/><br/> 32. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189-198.</p> <p class="reference"> 33. Taylor MA, Abrams R. The phenomenology of mania. A new look at some old patients. Arch Gen Psychiatry. 1973;29(4):520-522.<br/><br/> 34. Klerman GL. The spectrum of mania. Compr Psychiatry. 1981;22(1):11-20.<br/><br/> 35. Elias A, Thomas N, Sackeim HA. Electroconvulsive therapy in mania: a review of 80 years of clinical experience. Am J Psychiatry. 2021;178(3):229-239.<br/><br/> 36. Thom RP, Levy-Carrick NC, Bui M, et al. Delirium. Am J Psychiatry. 2019;176(10):785-793.<br/><br/> 37. Charlton BG, Kavanau JL. Delirium and psychotic symptoms--an integrative model. Med Hypotheses. 2002;58(1):24-27.<br/><br/> 38. Kramp P, Bolwig TG. Electroconvulsive therapy in acute delirious states. Compr Psychiatry. 1981;22(4):368-371.<br/><br/> 39. Mash DC. Excited delirium and sudden death: a syndromal disorder at the extreme end of the neuropsychiatric continuum. Front Physiol. 2016;7:435.<br/><br/> 40. Strawn JR, Keck PE Jr, Caroff SN. Neuroleptic malignant syndrome. Am J Psychiatry. 2007;164(6):870-876.<br/><br/> 41. Charney DS. Monoamine dysfunction and the pathophysiology and treatment of depression. J Clin Psychiatry. 1998;59 Suppl 14:11-14.<br/><br/> 42. Shiah IS, Yatham LN. Serotonin in mania and in the mechanism of action of mood stabilizers: a review of clinical studies. Bipolar Disord. 2000;2(2):77-92.<br/><br/> 43. Dalley JW, Roiser JP. Dopamine, serotonin and impulsivity. Neuroscience. 2012;215:42-58.<br/><br/> 44. Maldonado JR. Pathoetiological model of delirium: a comprehensive understanding of the neurobiology of delirium and an evidence-based approach to prevention and treatment. Crit Care Clin. 2008;24(4):789-856, ix.<br/><br/> 45. Maldonado JR. Neuropathogenesis of delirium: review of current etiologic theories and common pathways. Am J Geriatr Psychiatry. 2013;21(12):1190-1222.<br/><br/> 46. Rasmussen SA, Mazurek MF, Rosebush PI. Catatonia: our current understanding of its diagnosis, treatment and pathophysiology. World J Psychiatry. 2016;6(4):391-398.<br/><br/> 47. Northoff G, Steinke R, Czcervenka C, et al. Decreased density of GABA-A receptors in the left sensorimotor cortex in akinetic catatonia: investigation of in vivo benzodiazepine receptor binding. J Neurol Neurosurg Psychiatry. 1999;67(4):445-450.<br/><br/> 48. Daniels J. Catatonia: clinical aspects and neurobiological correlates. J Neuropsychiatry Clin Neurosci. 2009;21(4):371-380.<br/><br/> 49. Restrepo-Martínez M, Chacón-González J, Bayliss L, et al. Delirious mania as a neuropsychiatric presentation in patients with anti-N-methyl-D-aspartate receptor encephalitis. Psychosomatics. 2020;61(1):64-69.<br/><br/> 50. Dalmau J, Armangué T, Planagumà J, et al. An update on anti-NMDA receptor encephalitis for neurologists and psychiatrists: mechanisms and models. Lancet Neurol. 2019;18(11):1045-1057.<br/><br/> 51. Steardo L Jr, Steardo L, Verkhratsky A. Psychiatric face of COVID-19. Transl Psychiatry. 2020;10(1):261.<br/><br/> 52. Iqbal Y, Al Abdulla MA, Albrahim S, et al. Psychiatric presentation of patients with acute SARS-CoV-2 infection: a retrospective review of 50 consecutive patients seen by a consultation-liaison psychiatry team. BJPsych Open. 2020;6(5):e109.<br/><br/> 53. Gouse BM, Spears WE, Nieves Archibald A, et al. Catatonia in a hospitalized patient with COVID-19 and proposed immune-mediated mechanism. Brain Behav Immun. 2020;89:529-530.<br/><br/> 54. Caan MP, Lim CT, Howard M. A case of catatonia in a man with COVID-19. Psychosomatics. 2020;61(5):556-560.<br/><br/> 55. Zain SM, Muthukanagaraj P, Rahman N. Excited catatonia - a delayed neuropsychiatric complication of COVID-19 infection. Cureus. 2021;13(3):e13891.<br/><br/> 56. Chowdhury MA, Hossain N, Kashem MA, et al. Immune response in COVID-19: a review. J Infect Public Health. 2020;13(11):1619-1629.<br/><br/> 57. Radhakrishnan R, Kaser M, Guloksuz S. The link between the immune system, environment, and psychosis. Schizophr Bull. 2017;43(4):693-697.<br/><br/> 58. Fink M, Kellner CH, McCall WV. Optimizing ECT technique in treating catatonia. J ECT. 2016;32(3):149-150.<br/><br/> 59. Cohen WJ, Cohen NH. Lithium carbonate, haloperidol, and irreversible brain damage. JAMA. 1974;230(9):1283-1287.<br/><br/> 60. Davis MJ, de Nesnera A, Folks DG. Confused and nearly naked after going on spending sprees. Current Psychiatry. 2014;13(7):56-62.<br/><br/> 61. Stanley M, Chippa V, Aeddula NR, et al. <span class="iref">Rhabdomyolysis</span>. StatPearls Publishing; 2021.<br/><br/> 62. Fink M, Taylor MA. The catatonia syndrome: forgotten but not gone. Arch Gen Psychiatry. 2009;66(11):1173-1177.<br/><br/> 63. Hiremani RM, Thirthalli J, Tharayil BS, et al. Double-blind randomized controlled study comparing short-term efficacy of bifrontal and bitemporal electroconvulsive therapy in acute mania. Bipolar Disord. 2008;10(6):701-707.<br/><br/> 64. Barekatain M, Jahangard L, Haghighi M, et al. Bifrontal versus bitemporal electroconvulsive therapy in severe manic patients. J ECT. 2008;24(3):199-202.</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002644.SIG
Disable zoom
Off

Dear patients: Letters psychiatrists should and should not write

Article Type
Changed
Wed, 12/06/2023 - 10:09
Display Headline
Dear patients: Letters psychiatrists should and should not write

After several months of difficulty living in her current apartment complex, Ms. M asks you as her psychiatrist to write a letter to the management company requesting she be moved to an apartment on the opposite side of the maintenance closet because the noise aggravates her posttraumatic stress disorder. What should you consider when asked to write such a letter?

Psychiatric practice often extends beyond the treatment of mental illness to include addressing patients’ social well-being. Psychiatrists commonly inquire about a patient’s social situation to understand the impact of these environmental factors. Similarly, psychiatric illness may affect a patient’s ability to work or fulfill responsibilities. As a result, patients may ask their psychiatrists for assistance by requesting letters that address various aspects of their social well-being.1 These communications may address an array of topics, from a patient’s readiness to return to work to their ability to pay child support. This article focuses on the role psychiatrists have in writing patient-requested letters across a variety of topics, including the consideration of potential legal liability and ethical implications.

Types of letters

The categories of letters patients request can be divided into 2 groups. The first is comprised of letters relating to the patient’s medical needs (Table 12,3). These address the patient’s ability to work (eg, medical leave, return to work, or accommodations) or travel (eg, ability to drive or use public transportation), or need for specific medical treatment (ie, gender-affirming care or cannabis use in specific settings). The second group relates to legal requests such as excusal from jury duty, emotional support animals, or any other letter used specifically for legal purposes (in civil or criminal cases) (Table 21,4-6).

CP02212011_t1.png

The decision to write a letter on behalf of a patient should be based on whether you have sufficient knowledge to answer the referral question, and whether the requested evaluation fits within your role as the treating psychiatrist. Many requests fall short of the first condition. For example, a request to opine about an individual’s ability to perform their job duties requires specific knowledge and careful consideration of the patient’s work responsibilities, knowledge of the impact of their psychiatric symptoms, and specialized knowledge about interventions that would ameliorate symptoms in the specialized work setting. Most psychiatrists are not sufficiently familiar with a specific workplace to provide opinions regarding reasonable accommodations.

CP02212011_t2.png

The second condition refers to the role and responsibilities of the psychiatrist. Many letter requests are clearly within the scope of the clinical psychiatrist, such as a medical leave note due to a psychiatric decompensation or a jury duty excusal due to an unstable mental state. Other letters reach beyond the role of the general or treating psychiatrist, such as opinions about suitable housing or a patient’s competency to stand trial.

Components of letters

The decision to write or not to write a letter should be discussed with the patient. Identify the reasons for and against letter writing. If you decide to write a letter, the letter should have the following basic framework (Figure): the identity of the person who requested the letter, the referral question, and an answer to the referral question with a clear rationale. Describe the patient’s psychiatric diagnosis using DSM criteria. Any limitations to the answer should be identified. The letter should not go beyond the referral question and should not include information that was not requested. It also should be preserved in the medical record.

CP02212011_f1.png

It is recommended to write or review the letter in the presence of the patient to discuss the contents of the letter and what the psychiatrist can or cannot write. As in forensic reports, conclusory statements are not helpful. Provide descriptive information instead of relying on psychiatric jargon, and a rationale for the opinion as opposed to stating an opinion as fact. In the letter, you must acknowledge that your opinion is based upon information provided by the patient (and the patient’s family, when accurate) and as a result, is not fully objective.

Continue to: Liability and dual agency

 

 

Liability and dual agency

Psychiatrists are familiar with clinical situations in which a duty to the patient is mitigated or superseded by a duty to a third party. As the Tarasoff court famously stated, “the protective privilege ends where the public peril begins.”7

To be liable to either a patient or a third party means to be “bound or obliged in law or equity; responsible; chargeable; answerable; compellable to make satisfaction, compensation, or restitution.”8 Liabilities related to clinical treatment are well-established; medical students learn the fundamentals before ever treating a patient, and physicians carry malpractice insurance throughout their careers.

Less well-established is the liability a treating psychiatrist owes a third party when forming an opinion that impacts both their patient and the third party (eg, an employer when writing a return-to-work letter, or a disability insurer when qualifying a patient for disability benefits). The American Academy of Psychiatry and the Law discourages treating psychiatrists from performing these types of evaluations of their patients based on the inherent conflict of serving as a dual agent, or acting both as an advocate for the patient and as an independent evaluator striving for objectivity.9 However, such requests commonly arise, and some may be unavoidable.

Dual-agency situations subject the treating psychiatrist to avenues of legal action arising from the patient-doctor relationship as well as the forensic evaluator relationship. If a letter is written during a clinical treatment, all duties owed to the patient continue to apply, and the relevant benchmarks of local statutes and principle of a standard of care are relevant. It is conceivable that a patient could bring a negligence lawsuit based on a standard of care allegation (eg, that writing certain types of letters is so ordinary that failure to write them would fall below the standard of care). Confidentiality is also of the utmost importance,10 and you should obtain a written release of information from the patient before releasing any letter with privileged information about the patient.11 Additional relevant legal causes of action the patient could include are torts such as defamation of character, invasion of privacy, breach of contract, and intentional infliction of emotional distress. There is limited case law supporting patients’ rights to sue psychiatrists for defamation.10

A psychiatrist writing a letter to a third party may also subject themselves to avenues of legal action occurring outside the physician-patient relationship. Importantly, damages resulting from these breaches would not be covered by your malpractice insurance. Extreme cases involve allegations of fraud or perjury, which could be pursued in criminal court. If a psychiatrist intentionally deceives a third party for the purpose of obtaining some benefit for the patient, this is clear grounds for civil or criminal action. Fraud is defined as “a false representation of a matter of fact, whether by words or by conduct, by false or misleading allegations, or by concealment of that which should have been disclosed, which deceives and is intended to deceive another so that he shall act upon it to his legal injury.”8 Negligence can also be grounds for liability if a third party suffers injury or loss. Although the liability is clearer if the third party retains an independent psychiatrist rather than soliciting an opinion from a patient’s treating psychiatrist, both parties are subject to the claim of negligence.10

Continue to: There are some important protections...

 

 

There are some important protections that limit psychiatrists’ good-faith opinions from litigation. The primary one is the “professional medical judgment rule,” which shields physicians from the consequences of erroneous opinions so long as the examination was competent, complete, and performed in an ordinary fashion.10 In some cases, psychiatrists writing a letter or report for a government agency may also qualify for quasi-judicial immunity or witness immunity, but case law shows significant variation in when and how these privileges apply and whether such privileges would be applied to a clinical psychiatrist in the context of a traditional physician-patient relationship.12 In general, these privileges are not absolute and may not be sufficiently well-established to discourage a plaintiff from filing suit or prompt early judicial dismissal of a case.

Like all aspects of practicing medicine, letter writing is subject to scrutiny and accountability. Think carefully about your obligations and the potential consequences of writing or not writing a letter to a third party.

Ethical considerations

The decision to write a letter for a patient must be carefully considered from multiple angles.6 In addition to liability concerns, various ethical considerations also arise. Guided by the principles of beneficence, nonmaleficence, autonomy, and justice,13 we recommend the following approaches.

Maintain objectivity

During letter writing, a conflict of interest may arise between your allegiance to the patient and the imperative to provide accurate information.14-16 If the conflict is overwhelming, the most appropriate approach is to recuse yourself from the case and refer the patient to a third party. When electing to write a letter, you accept the responsibility to provide an objective assessment of the relevant situation. This promotes a just outcome and may also serve to promote the patient’s or society’s well-being.

Encourage activity and overall function

Evidence suggests that participation in multiple aspects of life promotes positive health outcomes.17,18 As a physician, it is your duty to promote health and support and facilitate accommodations that allow patients to participate and flourish in society. By the same logic, when approached by patients with a request for letters in support of reduced activity, you should consider not only the benefits but also the potential detriments of such disruptions. This may entail recommending temporary restrictions or modifications, as appropriate.

Continue to: Think beyond the patient

 

 

Think beyond the patient

Letter writing, particularly when recommending accommodations, can have implications beyond the patient.16 Such letters may cause unintended societal harm. For example, others may have to assume additional responsibilities; competitive goods (eg, housing) may be rendered to the patient rather than to a person with greater needs; and workplace safety could be compromised due to absence. Consider not only the individual patient but also possible public health and societal effects of letter writing.

Deciding not to write

From an ethical perspective, a physician cannot be compelled to write a letter if such an undertaking violates a stronger moral obligation. An example of this is if writing a letter could cause significant harm to the patient or society, or if writing a letter might compromise a physician’s professionalism.19 When you elect to not write a letter, the ethical principles of autonomy and truth telling dictate that you must inform your patients of this choice.6 You should also provide an explanation to the patient as long as such information would not cause undue psychological or physical harm.20,21

Schedule time to write letters

Some physicians implement policies that all letters are to be completed during scheduled appointments. Others designate administrative time to complete requested letters. Finally, some physicians flexibly complete such requests between appointments or during other undedicated time slots. Any of these approaches are justifiable, though some urgent requests may require more immediate attention outside of appointments. Some physicians may choose to bill for the letter writing if completed outside an appointment and the patient is treated in private practice. Whatever your policy, inform patients of it at the beginning of care and remind them when appropriate, such as before completing a letter that may be billed.

 

Manage uncertainty

Always strive for objectivity in letter writing. However, some requests inherently hinge on subjective reports and assessments. For example, a patient may request an excuse letter due to feeling unwell. In the absence of objective findings, what should you do? We advise the following.

Acquire collateral information. Adequate information is essential when making any medical recommendation. The same is true for writing letters. With the patient’s permission, you may need to contact relevant parties to better understand the circumstance or activity about which you are being asked to write a letter. For example, a patient may request leave from work due to injury. If the specific parameters of the work impeded by the injury are unclear to you, refrain from writing the letter and explain the rationale to the patient.

Continue to: Integrate prior knowledge of the patient

 

 

Integrate prior knowledge of the patient. No letter writing request exists in a vacuum. If you know the patient, the letter should be contextualized within the patient’s prior behaviors.

Stay within your scope

Given the various dilemmas and challenges, you may want to consider whether some letter writing is out of your professional scope.14-16 One solution would be to leave such requests to other entities (eg, requiring employers to retain medical personnel with specialized skills in occupational evaluations) and make such recommendations to patients. Regardless, physicians should think carefully about their professional boundaries and scope regarding letter requests and adopt and implement a consistent standard for all patients.

Regarding the letter requested by Ms. M, you should consider whether the appeal is consistent with the patient’s psychiatric illness. You should also consider whether you have sufficient knowledge about the patient’s living environment to support their claim. Such a letter should be written only if you understand both considerations. Regardless of your decision, you should explain your rationale to the patient.

 

Bottom Line

Patients may ask their psychiatrists to write letters that address aspects of their social well-being. However, psychiatrists must be alert to requests that are outside their scope of practice or ethically or legally fraught. Carefully consider whether writing a letter is appropriate and if not, discuss with the patient the reasons you cannot write such a letter and any recommended alternative avenues to address their request.

Related Resources

[embed:render:related:node:243699]

[embed:render:related:node:248104]

References

1. West S, Friedman SH. To be or not to be: treating psychiatrist and expert witness. Psychiatric Times. 2007;24(6). Accessed March 14, 2023. https://www.psychiatrictimes.com/view/be-or-not-be-treating-psychiatrist-and-expert-witness

2. Knoepflmacher D. ‘Medical necessity’ in psychiatry: whose definition is it anyway? Psychiatric News. 2016;51(18):12-14. https://psychnews.psychiatryonline.org/doi/10.1176/appi.pn.2016.9b14

3. Lampe JR. Recent developments in marijuana law (LSB10859). Congressional Research Service. 2022. Accessed October 25, 2023. https://crsreports.congress.gov/product/pdf/LSB/LSB10859/2

4. Brunnauer A, Buschert V, Segmiller F, et al. Mobility behaviour and driving status of patients with mental disorders – an exploratory study. Int J Psychiatry Clin Pract. 2016;20(1):40-46. doi:10.3109/13651501.2015.1089293

5. Chiu CW, Law CK, Cheng AS. Driver assessment service for people with mental illness. Hong Kong J Occup Ther. 2019;32(2):77-83. doi:10.1177/1569186119886773

6. Joshi KG. Service animals and emotional support animals: should you write that letter? Current Psychiatry. 2021;20(11):16-19. doi:10.12788/cp.0183

7. Tarasoff v Regents of University of California, 17 Cal 3d 425, 551 P2d 334, 131 Cal. Rptr. 14 (Cal 1976).

8. Black HC. Liability. Black’s Law Dictionary. Revised 4th ed. West Publishing; 1975:1060.

9. American Academy of Psychiatry and the Law. Ethics guidelines for the practice of forensic psychiatry. 2005. Accessed March 15, 2023. https://www.aapl.org/ethics.htm

10. Gold LH, Davidson JE. Do you understand your risk? Liability and third-party evaluations in civil litigation. J Am Acad Psychiatry Law. 2007;35(2):200-210.

11. Schouten R. Approach to the patient seeking disability benefits. In: Stern TA, Herman JB, Slavin PL, eds. The MGH Guide to Psychiatry in Primary Care. McGraw Hill; 1998:121-126.

12. Appelbaum PS. Law and psychiatry: liability for forensic evaluations: a word of caution. Psychiatr Serv. 2001;52(7):885-886. doi:10.1176/appi.ps.52.7.885

13. Varkey B. Principles of clinical ethics and their application to practice. Med Princ Pract. 2021;30(1):17-28. doi:10.1159/000509119

14. Mayhew HE, Nordlund DJ. Absenteeism certification: the physician’s role. J Fam Pract. 1988;26(6):651-655.

15. Younggren JN, Boisvert JA, Boness CL. Examining emotional support animals and role conflicts in professional psychology. Prof Psychol Res Pr. 2016;47(4):255-260. doi:10.1037/pro0000083

16. Carroll JD, Mohlenhoff BS, Kersten CM, et al. Laws and ethics related to emotional support animals. J Am Acad Psychiatry Law. 2020;48(4):509-518. doi:1-.29158/JAAPL.200047-20

17. Strully KW. Job loss and health in the U.S. labor market. Demography. 2009;46(2):221-246. doi:10.1353/dem.0.0050

18. Jurisic M, Bean M, Harbaugh J, et al. The personal physician’s role in helping patients with medical conditions stay at work or return to work. J Occup Environ Med. 2017;59(6):e125-131. doi:10.1097/JOM.0000000000001055

19. Munyaradzi M. Critical reflections on the principle of beneficence in biomedicine. Pan Afr Med J. 2012;11:29.

20. Beauchamp TL, Childress JF. Principles of Biomedical Ethics. 7th ed. Oxford University Press; 2012.

21. Gold M. Is honesty always the best policy? Ethical aspects of truth telling. Intern Med J. 2004;34(9-10):578-580. doi:10.1111/j.1445-5994.2004.00673.x

Article PDF
Author and Disclosure Information

Renée Sorrentino, MD, DFAPA
Clinical Assistant Professor, Part-time
Department of Psychiatry
Harvard Medical School
Boston, Massachusetts

Ren Belcher, MD
Lecturer in Psychiatry, Part-time
Harvard Medical School
Boston, Massachusetts

Rachel L. MacLean, MD
Clinical Fellow
Department of Psychiatry
Harvard Medical School
Boston, Massachusetts

Susan Hatters Friedman, MD, DFAPA
The Phillip J. Resnick Professor of Forensic Psychiatry
Professor of Psychiatry, Reproductive Biology, Pediatrics, and Law
Case Western Reserve University
Cleveland, Ohio

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Issue
Current Psychiatry - 22(12)
Publications
Page Number
11-15,21,24
Sections
Author and Disclosure Information

Renée Sorrentino, MD, DFAPA
Clinical Assistant Professor, Part-time
Department of Psychiatry
Harvard Medical School
Boston, Massachusetts

Ren Belcher, MD
Lecturer in Psychiatry, Part-time
Harvard Medical School
Boston, Massachusetts

Rachel L. MacLean, MD
Clinical Fellow
Department of Psychiatry
Harvard Medical School
Boston, Massachusetts

Susan Hatters Friedman, MD, DFAPA
The Phillip J. Resnick Professor of Forensic Psychiatry
Professor of Psychiatry, Reproductive Biology, Pediatrics, and Law
Case Western Reserve University
Cleveland, Ohio

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Author and Disclosure Information

Renée Sorrentino, MD, DFAPA
Clinical Assistant Professor, Part-time
Department of Psychiatry
Harvard Medical School
Boston, Massachusetts

Ren Belcher, MD
Lecturer in Psychiatry, Part-time
Harvard Medical School
Boston, Massachusetts

Rachel L. MacLean, MD
Clinical Fellow
Department of Psychiatry
Harvard Medical School
Boston, Massachusetts

Susan Hatters Friedman, MD, DFAPA
The Phillip J. Resnick Professor of Forensic Psychiatry
Professor of Psychiatry, Reproductive Biology, Pediatrics, and Law
Case Western Reserve University
Cleveland, Ohio

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Article PDF
Article PDF

After several months of difficulty living in her current apartment complex, Ms. M asks you as her psychiatrist to write a letter to the management company requesting she be moved to an apartment on the opposite side of the maintenance closet because the noise aggravates her posttraumatic stress disorder. What should you consider when asked to write such a letter?

Psychiatric practice often extends beyond the treatment of mental illness to include addressing patients’ social well-being. Psychiatrists commonly inquire about a patient’s social situation to understand the impact of these environmental factors. Similarly, psychiatric illness may affect a patient’s ability to work or fulfill responsibilities. As a result, patients may ask their psychiatrists for assistance by requesting letters that address various aspects of their social well-being.1 These communications may address an array of topics, from a patient’s readiness to return to work to their ability to pay child support. This article focuses on the role psychiatrists have in writing patient-requested letters across a variety of topics, including the consideration of potential legal liability and ethical implications.

Types of letters

The categories of letters patients request can be divided into 2 groups. The first is comprised of letters relating to the patient’s medical needs (Table 12,3). These address the patient’s ability to work (eg, medical leave, return to work, or accommodations) or travel (eg, ability to drive or use public transportation), or need for specific medical treatment (ie, gender-affirming care or cannabis use in specific settings). The second group relates to legal requests such as excusal from jury duty, emotional support animals, or any other letter used specifically for legal purposes (in civil or criminal cases) (Table 21,4-6).

CP02212011_t1.png

The decision to write a letter on behalf of a patient should be based on whether you have sufficient knowledge to answer the referral question, and whether the requested evaluation fits within your role as the treating psychiatrist. Many requests fall short of the first condition. For example, a request to opine about an individual’s ability to perform their job duties requires specific knowledge and careful consideration of the patient’s work responsibilities, knowledge of the impact of their psychiatric symptoms, and specialized knowledge about interventions that would ameliorate symptoms in the specialized work setting. Most psychiatrists are not sufficiently familiar with a specific workplace to provide opinions regarding reasonable accommodations.

CP02212011_t2.png

The second condition refers to the role and responsibilities of the psychiatrist. Many letter requests are clearly within the scope of the clinical psychiatrist, such as a medical leave note due to a psychiatric decompensation or a jury duty excusal due to an unstable mental state. Other letters reach beyond the role of the general or treating psychiatrist, such as opinions about suitable housing or a patient’s competency to stand trial.

Components of letters

The decision to write or not to write a letter should be discussed with the patient. Identify the reasons for and against letter writing. If you decide to write a letter, the letter should have the following basic framework (Figure): the identity of the person who requested the letter, the referral question, and an answer to the referral question with a clear rationale. Describe the patient’s psychiatric diagnosis using DSM criteria. Any limitations to the answer should be identified. The letter should not go beyond the referral question and should not include information that was not requested. It also should be preserved in the medical record.

CP02212011_f1.png

It is recommended to write or review the letter in the presence of the patient to discuss the contents of the letter and what the psychiatrist can or cannot write. As in forensic reports, conclusory statements are not helpful. Provide descriptive information instead of relying on psychiatric jargon, and a rationale for the opinion as opposed to stating an opinion as fact. In the letter, you must acknowledge that your opinion is based upon information provided by the patient (and the patient’s family, when accurate) and as a result, is not fully objective.

Continue to: Liability and dual agency

 

 

Liability and dual agency

Psychiatrists are familiar with clinical situations in which a duty to the patient is mitigated or superseded by a duty to a third party. As the Tarasoff court famously stated, “the protective privilege ends where the public peril begins.”7

To be liable to either a patient or a third party means to be “bound or obliged in law or equity; responsible; chargeable; answerable; compellable to make satisfaction, compensation, or restitution.”8 Liabilities related to clinical treatment are well-established; medical students learn the fundamentals before ever treating a patient, and physicians carry malpractice insurance throughout their careers.

Less well-established is the liability a treating psychiatrist owes a third party when forming an opinion that impacts both their patient and the third party (eg, an employer when writing a return-to-work letter, or a disability insurer when qualifying a patient for disability benefits). The American Academy of Psychiatry and the Law discourages treating psychiatrists from performing these types of evaluations of their patients based on the inherent conflict of serving as a dual agent, or acting both as an advocate for the patient and as an independent evaluator striving for objectivity.9 However, such requests commonly arise, and some may be unavoidable.

Dual-agency situations subject the treating psychiatrist to avenues of legal action arising from the patient-doctor relationship as well as the forensic evaluator relationship. If a letter is written during a clinical treatment, all duties owed to the patient continue to apply, and the relevant benchmarks of local statutes and principle of a standard of care are relevant. It is conceivable that a patient could bring a negligence lawsuit based on a standard of care allegation (eg, that writing certain types of letters is so ordinary that failure to write them would fall below the standard of care). Confidentiality is also of the utmost importance,10 and you should obtain a written release of information from the patient before releasing any letter with privileged information about the patient.11 Additional relevant legal causes of action the patient could include are torts such as defamation of character, invasion of privacy, breach of contract, and intentional infliction of emotional distress. There is limited case law supporting patients’ rights to sue psychiatrists for defamation.10

A psychiatrist writing a letter to a third party may also subject themselves to avenues of legal action occurring outside the physician-patient relationship. Importantly, damages resulting from these breaches would not be covered by your malpractice insurance. Extreme cases involve allegations of fraud or perjury, which could be pursued in criminal court. If a psychiatrist intentionally deceives a third party for the purpose of obtaining some benefit for the patient, this is clear grounds for civil or criminal action. Fraud is defined as “a false representation of a matter of fact, whether by words or by conduct, by false or misleading allegations, or by concealment of that which should have been disclosed, which deceives and is intended to deceive another so that he shall act upon it to his legal injury.”8 Negligence can also be grounds for liability if a third party suffers injury or loss. Although the liability is clearer if the third party retains an independent psychiatrist rather than soliciting an opinion from a patient’s treating psychiatrist, both parties are subject to the claim of negligence.10

Continue to: There are some important protections...

 

 

There are some important protections that limit psychiatrists’ good-faith opinions from litigation. The primary one is the “professional medical judgment rule,” which shields physicians from the consequences of erroneous opinions so long as the examination was competent, complete, and performed in an ordinary fashion.10 In some cases, psychiatrists writing a letter or report for a government agency may also qualify for quasi-judicial immunity or witness immunity, but case law shows significant variation in when and how these privileges apply and whether such privileges would be applied to a clinical psychiatrist in the context of a traditional physician-patient relationship.12 In general, these privileges are not absolute and may not be sufficiently well-established to discourage a plaintiff from filing suit or prompt early judicial dismissal of a case.

Like all aspects of practicing medicine, letter writing is subject to scrutiny and accountability. Think carefully about your obligations and the potential consequences of writing or not writing a letter to a third party.

Ethical considerations

The decision to write a letter for a patient must be carefully considered from multiple angles.6 In addition to liability concerns, various ethical considerations also arise. Guided by the principles of beneficence, nonmaleficence, autonomy, and justice,13 we recommend the following approaches.

Maintain objectivity

During letter writing, a conflict of interest may arise between your allegiance to the patient and the imperative to provide accurate information.14-16 If the conflict is overwhelming, the most appropriate approach is to recuse yourself from the case and refer the patient to a third party. When electing to write a letter, you accept the responsibility to provide an objective assessment of the relevant situation. This promotes a just outcome and may also serve to promote the patient’s or society’s well-being.

Encourage activity and overall function

Evidence suggests that participation in multiple aspects of life promotes positive health outcomes.17,18 As a physician, it is your duty to promote health and support and facilitate accommodations that allow patients to participate and flourish in society. By the same logic, when approached by patients with a request for letters in support of reduced activity, you should consider not only the benefits but also the potential detriments of such disruptions. This may entail recommending temporary restrictions or modifications, as appropriate.

Continue to: Think beyond the patient

 

 

Think beyond the patient

Letter writing, particularly when recommending accommodations, can have implications beyond the patient.16 Such letters may cause unintended societal harm. For example, others may have to assume additional responsibilities; competitive goods (eg, housing) may be rendered to the patient rather than to a person with greater needs; and workplace safety could be compromised due to absence. Consider not only the individual patient but also possible public health and societal effects of letter writing.

Deciding not to write

From an ethical perspective, a physician cannot be compelled to write a letter if such an undertaking violates a stronger moral obligation. An example of this is if writing a letter could cause significant harm to the patient or society, or if writing a letter might compromise a physician’s professionalism.19 When you elect to not write a letter, the ethical principles of autonomy and truth telling dictate that you must inform your patients of this choice.6 You should also provide an explanation to the patient as long as such information would not cause undue psychological or physical harm.20,21

Schedule time to write letters

Some physicians implement policies that all letters are to be completed during scheduled appointments. Others designate administrative time to complete requested letters. Finally, some physicians flexibly complete such requests between appointments or during other undedicated time slots. Any of these approaches are justifiable, though some urgent requests may require more immediate attention outside of appointments. Some physicians may choose to bill for the letter writing if completed outside an appointment and the patient is treated in private practice. Whatever your policy, inform patients of it at the beginning of care and remind them when appropriate, such as before completing a letter that may be billed.

 

Manage uncertainty

Always strive for objectivity in letter writing. However, some requests inherently hinge on subjective reports and assessments. For example, a patient may request an excuse letter due to feeling unwell. In the absence of objective findings, what should you do? We advise the following.

Acquire collateral information. Adequate information is essential when making any medical recommendation. The same is true for writing letters. With the patient’s permission, you may need to contact relevant parties to better understand the circumstance or activity about which you are being asked to write a letter. For example, a patient may request leave from work due to injury. If the specific parameters of the work impeded by the injury are unclear to you, refrain from writing the letter and explain the rationale to the patient.

Continue to: Integrate prior knowledge of the patient

 

 

Integrate prior knowledge of the patient. No letter writing request exists in a vacuum. If you know the patient, the letter should be contextualized within the patient’s prior behaviors.

Stay within your scope

Given the various dilemmas and challenges, you may want to consider whether some letter writing is out of your professional scope.14-16 One solution would be to leave such requests to other entities (eg, requiring employers to retain medical personnel with specialized skills in occupational evaluations) and make such recommendations to patients. Regardless, physicians should think carefully about their professional boundaries and scope regarding letter requests and adopt and implement a consistent standard for all patients.

Regarding the letter requested by Ms. M, you should consider whether the appeal is consistent with the patient’s psychiatric illness. You should also consider whether you have sufficient knowledge about the patient’s living environment to support their claim. Such a letter should be written only if you understand both considerations. Regardless of your decision, you should explain your rationale to the patient.

 

Bottom Line

Patients may ask their psychiatrists to write letters that address aspects of their social well-being. However, psychiatrists must be alert to requests that are outside their scope of practice or ethically or legally fraught. Carefully consider whether writing a letter is appropriate and if not, discuss with the patient the reasons you cannot write such a letter and any recommended alternative avenues to address their request.

Related Resources

[embed:render:related:node:243699]

[embed:render:related:node:248104]

After several months of difficulty living in her current apartment complex, Ms. M asks you as her psychiatrist to write a letter to the management company requesting she be moved to an apartment on the opposite side of the maintenance closet because the noise aggravates her posttraumatic stress disorder. What should you consider when asked to write such a letter?

Psychiatric practice often extends beyond the treatment of mental illness to include addressing patients’ social well-being. Psychiatrists commonly inquire about a patient’s social situation to understand the impact of these environmental factors. Similarly, psychiatric illness may affect a patient’s ability to work or fulfill responsibilities. As a result, patients may ask their psychiatrists for assistance by requesting letters that address various aspects of their social well-being.1 These communications may address an array of topics, from a patient’s readiness to return to work to their ability to pay child support. This article focuses on the role psychiatrists have in writing patient-requested letters across a variety of topics, including the consideration of potential legal liability and ethical implications.

Types of letters

The categories of letters patients request can be divided into 2 groups. The first is comprised of letters relating to the patient’s medical needs (Table 12,3). These address the patient’s ability to work (eg, medical leave, return to work, or accommodations) or travel (eg, ability to drive or use public transportation), or need for specific medical treatment (ie, gender-affirming care or cannabis use in specific settings). The second group relates to legal requests such as excusal from jury duty, emotional support animals, or any other letter used specifically for legal purposes (in civil or criminal cases) (Table 21,4-6).

CP02212011_t1.png

The decision to write a letter on behalf of a patient should be based on whether you have sufficient knowledge to answer the referral question, and whether the requested evaluation fits within your role as the treating psychiatrist. Many requests fall short of the first condition. For example, a request to opine about an individual’s ability to perform their job duties requires specific knowledge and careful consideration of the patient’s work responsibilities, knowledge of the impact of their psychiatric symptoms, and specialized knowledge about interventions that would ameliorate symptoms in the specialized work setting. Most psychiatrists are not sufficiently familiar with a specific workplace to provide opinions regarding reasonable accommodations.

CP02212011_t2.png

The second condition refers to the role and responsibilities of the psychiatrist. Many letter requests are clearly within the scope of the clinical psychiatrist, such as a medical leave note due to a psychiatric decompensation or a jury duty excusal due to an unstable mental state. Other letters reach beyond the role of the general or treating psychiatrist, such as opinions about suitable housing or a patient’s competency to stand trial.

Components of letters

The decision to write or not to write a letter should be discussed with the patient. Identify the reasons for and against letter writing. If you decide to write a letter, the letter should have the following basic framework (Figure): the identity of the person who requested the letter, the referral question, and an answer to the referral question with a clear rationale. Describe the patient’s psychiatric diagnosis using DSM criteria. Any limitations to the answer should be identified. The letter should not go beyond the referral question and should not include information that was not requested. It also should be preserved in the medical record.

CP02212011_f1.png

It is recommended to write or review the letter in the presence of the patient to discuss the contents of the letter and what the psychiatrist can or cannot write. As in forensic reports, conclusory statements are not helpful. Provide descriptive information instead of relying on psychiatric jargon, and a rationale for the opinion as opposed to stating an opinion as fact. In the letter, you must acknowledge that your opinion is based upon information provided by the patient (and the patient’s family, when accurate) and as a result, is not fully objective.

Continue to: Liability and dual agency

 

 

Liability and dual agency

Psychiatrists are familiar with clinical situations in which a duty to the patient is mitigated or superseded by a duty to a third party. As the Tarasoff court famously stated, “the protective privilege ends where the public peril begins.”7

To be liable to either a patient or a third party means to be “bound or obliged in law or equity; responsible; chargeable; answerable; compellable to make satisfaction, compensation, or restitution.”8 Liabilities related to clinical treatment are well-established; medical students learn the fundamentals before ever treating a patient, and physicians carry malpractice insurance throughout their careers.

Less well-established is the liability a treating psychiatrist owes a third party when forming an opinion that impacts both their patient and the third party (eg, an employer when writing a return-to-work letter, or a disability insurer when qualifying a patient for disability benefits). The American Academy of Psychiatry and the Law discourages treating psychiatrists from performing these types of evaluations of their patients based on the inherent conflict of serving as a dual agent, or acting both as an advocate for the patient and as an independent evaluator striving for objectivity.9 However, such requests commonly arise, and some may be unavoidable.

Dual-agency situations subject the treating psychiatrist to avenues of legal action arising from the patient-doctor relationship as well as the forensic evaluator relationship. If a letter is written during a clinical treatment, all duties owed to the patient continue to apply, and the relevant benchmarks of local statutes and principle of a standard of care are relevant. It is conceivable that a patient could bring a negligence lawsuit based on a standard of care allegation (eg, that writing certain types of letters is so ordinary that failure to write them would fall below the standard of care). Confidentiality is also of the utmost importance,10 and you should obtain a written release of information from the patient before releasing any letter with privileged information about the patient.11 Additional relevant legal causes of action the patient could include are torts such as defamation of character, invasion of privacy, breach of contract, and intentional infliction of emotional distress. There is limited case law supporting patients’ rights to sue psychiatrists for defamation.10

A psychiatrist writing a letter to a third party may also subject themselves to avenues of legal action occurring outside the physician-patient relationship. Importantly, damages resulting from these breaches would not be covered by your malpractice insurance. Extreme cases involve allegations of fraud or perjury, which could be pursued in criminal court. If a psychiatrist intentionally deceives a third party for the purpose of obtaining some benefit for the patient, this is clear grounds for civil or criminal action. Fraud is defined as “a false representation of a matter of fact, whether by words or by conduct, by false or misleading allegations, or by concealment of that which should have been disclosed, which deceives and is intended to deceive another so that he shall act upon it to his legal injury.”8 Negligence can also be grounds for liability if a third party suffers injury or loss. Although the liability is clearer if the third party retains an independent psychiatrist rather than soliciting an opinion from a patient’s treating psychiatrist, both parties are subject to the claim of negligence.10

Continue to: There are some important protections...

 

 

There are some important protections that limit psychiatrists’ good-faith opinions from litigation. The primary one is the “professional medical judgment rule,” which shields physicians from the consequences of erroneous opinions so long as the examination was competent, complete, and performed in an ordinary fashion.10 In some cases, psychiatrists writing a letter or report for a government agency may also qualify for quasi-judicial immunity or witness immunity, but case law shows significant variation in when and how these privileges apply and whether such privileges would be applied to a clinical psychiatrist in the context of a traditional physician-patient relationship.12 In general, these privileges are not absolute and may not be sufficiently well-established to discourage a plaintiff from filing suit or prompt early judicial dismissal of a case.

Like all aspects of practicing medicine, letter writing is subject to scrutiny and accountability. Think carefully about your obligations and the potential consequences of writing or not writing a letter to a third party.

Ethical considerations

The decision to write a letter for a patient must be carefully considered from multiple angles.6 In addition to liability concerns, various ethical considerations also arise. Guided by the principles of beneficence, nonmaleficence, autonomy, and justice,13 we recommend the following approaches.

Maintain objectivity

During letter writing, a conflict of interest may arise between your allegiance to the patient and the imperative to provide accurate information.14-16 If the conflict is overwhelming, the most appropriate approach is to recuse yourself from the case and refer the patient to a third party. When electing to write a letter, you accept the responsibility to provide an objective assessment of the relevant situation. This promotes a just outcome and may also serve to promote the patient’s or society’s well-being.

Encourage activity and overall function

Evidence suggests that participation in multiple aspects of life promotes positive health outcomes.17,18 As a physician, it is your duty to promote health and support and facilitate accommodations that allow patients to participate and flourish in society. By the same logic, when approached by patients with a request for letters in support of reduced activity, you should consider not only the benefits but also the potential detriments of such disruptions. This may entail recommending temporary restrictions or modifications, as appropriate.

Continue to: Think beyond the patient

 

 

Think beyond the patient

Letter writing, particularly when recommending accommodations, can have implications beyond the patient.16 Such letters may cause unintended societal harm. For example, others may have to assume additional responsibilities; competitive goods (eg, housing) may be rendered to the patient rather than to a person with greater needs; and workplace safety could be compromised due to absence. Consider not only the individual patient but also possible public health and societal effects of letter writing.

Deciding not to write

From an ethical perspective, a physician cannot be compelled to write a letter if such an undertaking violates a stronger moral obligation. An example of this is if writing a letter could cause significant harm to the patient or society, or if writing a letter might compromise a physician’s professionalism.19 When you elect to not write a letter, the ethical principles of autonomy and truth telling dictate that you must inform your patients of this choice.6 You should also provide an explanation to the patient as long as such information would not cause undue psychological or physical harm.20,21

Schedule time to write letters

Some physicians implement policies that all letters are to be completed during scheduled appointments. Others designate administrative time to complete requested letters. Finally, some physicians flexibly complete such requests between appointments or during other undedicated time slots. Any of these approaches are justifiable, though some urgent requests may require more immediate attention outside of appointments. Some physicians may choose to bill for the letter writing if completed outside an appointment and the patient is treated in private practice. Whatever your policy, inform patients of it at the beginning of care and remind them when appropriate, such as before completing a letter that may be billed.

 

Manage uncertainty

Always strive for objectivity in letter writing. However, some requests inherently hinge on subjective reports and assessments. For example, a patient may request an excuse letter due to feeling unwell. In the absence of objective findings, what should you do? We advise the following.

Acquire collateral information. Adequate information is essential when making any medical recommendation. The same is true for writing letters. With the patient’s permission, you may need to contact relevant parties to better understand the circumstance or activity about which you are being asked to write a letter. For example, a patient may request leave from work due to injury. If the specific parameters of the work impeded by the injury are unclear to you, refrain from writing the letter and explain the rationale to the patient.

Continue to: Integrate prior knowledge of the patient

 

 

Integrate prior knowledge of the patient. No letter writing request exists in a vacuum. If you know the patient, the letter should be contextualized within the patient’s prior behaviors.

Stay within your scope

Given the various dilemmas and challenges, you may want to consider whether some letter writing is out of your professional scope.14-16 One solution would be to leave such requests to other entities (eg, requiring employers to retain medical personnel with specialized skills in occupational evaluations) and make such recommendations to patients. Regardless, physicians should think carefully about their professional boundaries and scope regarding letter requests and adopt and implement a consistent standard for all patients.

Regarding the letter requested by Ms. M, you should consider whether the appeal is consistent with the patient’s psychiatric illness. You should also consider whether you have sufficient knowledge about the patient’s living environment to support their claim. Such a letter should be written only if you understand both considerations. Regardless of your decision, you should explain your rationale to the patient.

 

Bottom Line

Patients may ask their psychiatrists to write letters that address aspects of their social well-being. However, psychiatrists must be alert to requests that are outside their scope of practice or ethically or legally fraught. Carefully consider whether writing a letter is appropriate and if not, discuss with the patient the reasons you cannot write such a letter and any recommended alternative avenues to address their request.

Related Resources

[embed:render:related:node:243699]

[embed:render:related:node:248104]

References

1. West S, Friedman SH. To be or not to be: treating psychiatrist and expert witness. Psychiatric Times. 2007;24(6). Accessed March 14, 2023. https://www.psychiatrictimes.com/view/be-or-not-be-treating-psychiatrist-and-expert-witness

2. Knoepflmacher D. ‘Medical necessity’ in psychiatry: whose definition is it anyway? Psychiatric News. 2016;51(18):12-14. https://psychnews.psychiatryonline.org/doi/10.1176/appi.pn.2016.9b14

3. Lampe JR. Recent developments in marijuana law (LSB10859). Congressional Research Service. 2022. Accessed October 25, 2023. https://crsreports.congress.gov/product/pdf/LSB/LSB10859/2

4. Brunnauer A, Buschert V, Segmiller F, et al. Mobility behaviour and driving status of patients with mental disorders – an exploratory study. Int J Psychiatry Clin Pract. 2016;20(1):40-46. doi:10.3109/13651501.2015.1089293

5. Chiu CW, Law CK, Cheng AS. Driver assessment service for people with mental illness. Hong Kong J Occup Ther. 2019;32(2):77-83. doi:10.1177/1569186119886773

6. Joshi KG. Service animals and emotional support animals: should you write that letter? Current Psychiatry. 2021;20(11):16-19. doi:10.12788/cp.0183

7. Tarasoff v Regents of University of California, 17 Cal 3d 425, 551 P2d 334, 131 Cal. Rptr. 14 (Cal 1976).

8. Black HC. Liability. Black’s Law Dictionary. Revised 4th ed. West Publishing; 1975:1060.

9. American Academy of Psychiatry and the Law. Ethics guidelines for the practice of forensic psychiatry. 2005. Accessed March 15, 2023. https://www.aapl.org/ethics.htm

10. Gold LH, Davidson JE. Do you understand your risk? Liability and third-party evaluations in civil litigation. J Am Acad Psychiatry Law. 2007;35(2):200-210.

11. Schouten R. Approach to the patient seeking disability benefits. In: Stern TA, Herman JB, Slavin PL, eds. The MGH Guide to Psychiatry in Primary Care. McGraw Hill; 1998:121-126.

12. Appelbaum PS. Law and psychiatry: liability for forensic evaluations: a word of caution. Psychiatr Serv. 2001;52(7):885-886. doi:10.1176/appi.ps.52.7.885

13. Varkey B. Principles of clinical ethics and their application to practice. Med Princ Pract. 2021;30(1):17-28. doi:10.1159/000509119

14. Mayhew HE, Nordlund DJ. Absenteeism certification: the physician’s role. J Fam Pract. 1988;26(6):651-655.

15. Younggren JN, Boisvert JA, Boness CL. Examining emotional support animals and role conflicts in professional psychology. Prof Psychol Res Pr. 2016;47(4):255-260. doi:10.1037/pro0000083

16. Carroll JD, Mohlenhoff BS, Kersten CM, et al. Laws and ethics related to emotional support animals. J Am Acad Psychiatry Law. 2020;48(4):509-518. doi:1-.29158/JAAPL.200047-20

17. Strully KW. Job loss and health in the U.S. labor market. Demography. 2009;46(2):221-246. doi:10.1353/dem.0.0050

18. Jurisic M, Bean M, Harbaugh J, et al. The personal physician’s role in helping patients with medical conditions stay at work or return to work. J Occup Environ Med. 2017;59(6):e125-131. doi:10.1097/JOM.0000000000001055

19. Munyaradzi M. Critical reflections on the principle of beneficence in biomedicine. Pan Afr Med J. 2012;11:29.

20. Beauchamp TL, Childress JF. Principles of Biomedical Ethics. 7th ed. Oxford University Press; 2012.

21. Gold M. Is honesty always the best policy? Ethical aspects of truth telling. Intern Med J. 2004;34(9-10):578-580. doi:10.1111/j.1445-5994.2004.00673.x

References

1. West S, Friedman SH. To be or not to be: treating psychiatrist and expert witness. Psychiatric Times. 2007;24(6). Accessed March 14, 2023. https://www.psychiatrictimes.com/view/be-or-not-be-treating-psychiatrist-and-expert-witness

2. Knoepflmacher D. ‘Medical necessity’ in psychiatry: whose definition is it anyway? Psychiatric News. 2016;51(18):12-14. https://psychnews.psychiatryonline.org/doi/10.1176/appi.pn.2016.9b14

3. Lampe JR. Recent developments in marijuana law (LSB10859). Congressional Research Service. 2022. Accessed October 25, 2023. https://crsreports.congress.gov/product/pdf/LSB/LSB10859/2

4. Brunnauer A, Buschert V, Segmiller F, et al. Mobility behaviour and driving status of patients with mental disorders – an exploratory study. Int J Psychiatry Clin Pract. 2016;20(1):40-46. doi:10.3109/13651501.2015.1089293

5. Chiu CW, Law CK, Cheng AS. Driver assessment service for people with mental illness. Hong Kong J Occup Ther. 2019;32(2):77-83. doi:10.1177/1569186119886773

6. Joshi KG. Service animals and emotional support animals: should you write that letter? Current Psychiatry. 2021;20(11):16-19. doi:10.12788/cp.0183

7. Tarasoff v Regents of University of California, 17 Cal 3d 425, 551 P2d 334, 131 Cal. Rptr. 14 (Cal 1976).

8. Black HC. Liability. Black’s Law Dictionary. Revised 4th ed. West Publishing; 1975:1060.

9. American Academy of Psychiatry and the Law. Ethics guidelines for the practice of forensic psychiatry. 2005. Accessed March 15, 2023. https://www.aapl.org/ethics.htm

10. Gold LH, Davidson JE. Do you understand your risk? Liability and third-party evaluations in civil litigation. J Am Acad Psychiatry Law. 2007;35(2):200-210.

11. Schouten R. Approach to the patient seeking disability benefits. In: Stern TA, Herman JB, Slavin PL, eds. The MGH Guide to Psychiatry in Primary Care. McGraw Hill; 1998:121-126.

12. Appelbaum PS. Law and psychiatry: liability for forensic evaluations: a word of caution. Psychiatr Serv. 2001;52(7):885-886. doi:10.1176/appi.ps.52.7.885

13. Varkey B. Principles of clinical ethics and their application to practice. Med Princ Pract. 2021;30(1):17-28. doi:10.1159/000509119

14. Mayhew HE, Nordlund DJ. Absenteeism certification: the physician’s role. J Fam Pract. 1988;26(6):651-655.

15. Younggren JN, Boisvert JA, Boness CL. Examining emotional support animals and role conflicts in professional psychology. Prof Psychol Res Pr. 2016;47(4):255-260. doi:10.1037/pro0000083

16. Carroll JD, Mohlenhoff BS, Kersten CM, et al. Laws and ethics related to emotional support animals. J Am Acad Psychiatry Law. 2020;48(4):509-518. doi:1-.29158/JAAPL.200047-20

17. Strully KW. Job loss and health in the U.S. labor market. Demography. 2009;46(2):221-246. doi:10.1353/dem.0.0050

18. Jurisic M, Bean M, Harbaugh J, et al. The personal physician’s role in helping patients with medical conditions stay at work or return to work. J Occup Environ Med. 2017;59(6):e125-131. doi:10.1097/JOM.0000000000001055

19. Munyaradzi M. Critical reflections on the principle of beneficence in biomedicine. Pan Afr Med J. 2012;11:29.

20. Beauchamp TL, Childress JF. Principles of Biomedical Ethics. 7th ed. Oxford University Press; 2012.

21. Gold M. Is honesty always the best policy? Ethical aspects of truth telling. Intern Med J. 2004;34(9-10):578-580. doi:10.1111/j.1445-5994.2004.00673.x

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
11-15,21,24
Page Number
11-15,21,24
Publications
Publications
Article Type
Display Headline
Dear patients: Letters psychiatrists should and should not write
Display Headline
Dear patients: Letters psychiatrists should and should not write
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1223 Sorrentino</fileName> <TBEID>0C02E915.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02E915</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Dear patients: Letters psychiatr</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231129T080212</firstPublished> <LastPublished>20231129T080212</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231129T080211</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>R&#13;enée Sorrentino, MD, DFAPA, Ren Belcher, MD</byline> <bylineText/> <bylineFull>R&#13;enée Sorrentino, MD, DFAPA, Ren Belcher, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>11-15,21,24</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>After several months of difficulty living in her current apartment complex, Ms. M asks you as her psychiatrist to write a letter to the management company reque</metaDescription> <articlePDF>299323</articlePDF> <teaserImage/> <title>Dear patients: Letters psychiatrists should and should not write</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords> <keyword>medicolegal</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">91</term> </sections> <topics> <term canonical="true">278</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002653.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Dear patients: Letters psychiatrists should and should not write</title> <deck/> </itemMeta> <itemContent> <p class="abstract">How to determine if writing the letter your patient asked you for is ethical and appropriate</p> <p>After several months of difficulty living in her current apartment complex, Ms. M asks you as her psychiatrist to write a letter to the management company requesting she be moved to an apartment on the opposite side of the maintenance closet because the noise aggravates her posttraumatic stress disorder. What should you consider when asked to write such a letter?</p> <p><br/><br/>Psychiatric practice often extends beyond the treatment of mental illness to include addressing patients’ social well-being. Psychiatrists commonly inquire about a patient’s social situation to understand the impact of these environmental factors. Similarly, psychiatric illness may affect a patient’s ability to work or fulfill responsibilities. As a result, patients may ask their psychiatrists for assistance by requesting letters that address various aspects of their social well-being.<sup>1</sup> These communications may address an array of topics, from a patient’s readiness to return to work to their ability to pay child support. This article focuses on the role psychiatrists have in writing patient-requested letters across a variety of topics, including the consideration of potential legal liability and ethical implications.</p> <h3>Types of letters</h3> <p>The categories of letters patients request can be divided into 2 groups. The first is comprised of letters relating to the patient’s medical needs (<span class="bitalic">Table 1</span><sup>2,3</sup>). These address the patient’s ability to work (eg, medical leave, return to work, or accommodations) or travel (eg, ability to drive or use public transportation), or need for specific medical treatment (ie, gender-affirming care or cannabis use in specific settings). The second group relates to legal requests such as excusal from jury duty, emotional support animals, or any other letter used specifically for legal purposes (in civil or criminal cases) (<span class="bitalic">Table 2</span>,<sup>1,4-6</sup> <span class="bitalic">page 14</span>).</p> <p>The decision to write a letter on behalf of a patient should be based on whether you have sufficient knowledge to answer the referral question, and whether the requested evaluation fits within your role as the treating psychiatrist. Many requests fall short of the first condition. For example, a request to opine about an individual’s ability to perform their job duties requires specific knowledge and careful consideration of the patient’s work responsibilities, knowledge of the impact of their psychiatric symptoms, and specialized knowledge about interventions that would ameliorate symptoms in the specialized work setting. Most psychiatrists are not sufficiently familiar with a specific workplace to provide opinions regarding reasonable accommodations.<br/><br/>The second condition refers to the role and responsibilities of the psychiatrist. Many letter requests are clearly within the scope of the clinical psychiatrist, such as a medical leave note due to a psychiatric decompensation or a jury duty excusal due to an unstable mental state. Other letters reach beyond the role of the general or treating psychiatrist, such as opinions about suitable housing or a patient’s competency to stand trial.</p> <h3>Components of letters</h3> <p>The decision to write or not to write a letter should be discussed with the patient. Identify the reasons for and against letter writing. If you decide to write a letter, the letter should have the following basic framework (<b><i>Figure</i></b>, <b><i>page 15</i></b>): the identity of the person who requested the letter, the referral question, and an answer to the referral question with a clear rationale. Describe the patient’s psychiatric diagnosis using DSM criteria. Any limitations to the answer should be identified. The letter should not go beyond the referral question and should not include information that was not requested. It also should be preserved in the medical record.</p> <p>It is recommended to write or review the letter in the presence of the patient to discuss the contents of the letter and what the psychiatrist can or cannot write. As in forensic reports, conclusory statements are not helpful. Provide descriptive information instead of relying on psychiatric jargon, and a rationale for the opinion as opposed to stating an opinion as fact. In the letter, you must acknowledge that your opinion is based upon information provided by the patient (and the patient’s family, when accurate) and as a result, is not fully objective.</p> <h3>Liability and dual agency</h3> <p>Psychiatrists are familiar with clinical situations in which a duty to the patient is mitigated or superseded by a duty to a third party. As the Tarasoff court famously stated, “the protective privilege ends where the public peril begins.”<sup>7</sup></p> <p>To be liable to either a patient or a third party means to be “bound or obliged in law or equity; responsible; chargeable; answerable; compellable to make satisfaction, compensation, or restitution.”<sup>8</sup> Liabilities related to clinical treatment are well-established; medical students learn the fundamentals before ever treating a patient, and physicians carry malpractice insurance throughout their careers.<br/><br/>Less well-established is the liability a treating psychiatrist owes a third party when forming an opinion that impacts both their patient and the third party (eg, an employer when writing a return-to-work letter, or a disability insurer when qualifying a patient for disability benefits). The American Academy of Psychiatry and the Law discourages treating psychiatrists from performing these types of evaluations of their patients based on the inherent conflict of serving as a dual agent, or acting both as an advocate for the patient and as an independent evaluator striving for objectivity.<sup>9</sup> However, such requests commonly arise, and some may be unavoidable.<br/><br/>Dual-agency situations subject the treating psychiatrist to avenues of legal action arising from the patient-doctor relationship as well as the forensic evaluator relationship. If a letter is written during a clinical treatment, all duties owed to the patient continue to apply, and the relevant benchmarks of local statutes and principle of a standard of care are relevant. It is conceivable that a patient could bring a negligence lawsuit based on a standard of care allegation (eg, that writing certain types of letters is so ordinary that failure to write them would fall below the standard of care). Confidentiality is also of the utmost importance,<sup>10</sup> and you should obtain a written release of information from the patient before releasing any letter with privileged information about the patient.<sup>11</sup> Additional relevant legal causes of action the patient could include are torts such as defamation of character, invasion of privacy, breach of contract, and intentional infliction of emotional distress. There is limited case law supporting patients’ rights to sue psychiatrists for defamation.<sup>10</sup><br/><br/>A psychiatrist writing a letter to a third party may also subject themselves to avenues of legal action occurring outside the physician-patient relationship. Importantly, damages resulting from these breaches would not be covered by your malpractice insurance. Extreme cases involve allegations of fraud or perjury, which could be pursued in criminal court. If a psychiatrist intentionally deceives a third party for the purpose of obtaining some benefit for the patient, this is clear grounds for civil or criminal action. Fraud is defined as “a false representation of a matter of fact, whether by words or by conduct, by false or misleading allegations, or by concealment of that which should have been disclosed, which deceives and is intended to deceive another so that he shall act upon it to his legal injury.”<sup>8</sup> Negligence can also be grounds for liability if a third party suffers injury or loss. Although the liability is clearer if the third party retains an independent psychiatrist rather than soliciting an opinion from a patient’s treating psychiatrist, both parties are subject to the claim of negligence.<sup>10<br/><br/></sup>There are some important protections that limit psychiatrists’ good-faith opinions from litigation. The primary one is the “professional medical judgment rule,” which shields physicians from the consequences of erroneous opinions so long as the examination was competent, complete, and performed in an ordinary fashion.<sup>10</sup> In some cases, psychiatrists writing a letter or report for a government agency may also qualify for quasi-judicial immunity or witness immunity, but case law shows significant variation in when and how these privileges apply and whether such privileges would be applied to a clinical psychiatrist in the context of a traditional physician-patient relationship.<sup>12</sup> In general, these privileges are not absolute and may not be sufficiently well-established to discourage a plaintiff from filing suit or prompt early judicial dismissal of a case.<br/><br/>Like all aspects of practicing medicine, letter writing is subject to scrutiny and accountability. Think carefully about your obligations and the potential consequences of writing or not writing a letter to a third party.</p> <h3>Ethical considerations</h3> <p>The decision to write a letter for a patient must be carefully considered from multiple angles.<sup>6</sup> In addition to liability concerns, various ethical considerations also arise. Guided by the principles of beneficence, nonmaleficence, autonomy, and justice,<sup>13</sup> we recommend the following approaches.<br/><br/><span class="bbody"><br/><br/>Maintain objectivity</span>During letter writing, a conflict of interest may arise between your allegiance to the patient and the imperative to provide accurate information.<sup>14-16</sup> If the conflict is overwhelming, the most appropriate approach is to recuse yourself from the case and refer the patient to a third party. When electing to write a letter, you accept the responsibility to provide an objective assessment of the relevant situation. This promotes a just outcome and may also serve to promote the patient’s or society’s well-being.<br/><br/><span class="bbody">Encourage activity and overall function</span>Evidence suggests that participation in multiple aspects of life promotes positive health outcomes.<sup>17,18</sup> As a physician, it is your duty to promote health and support and facilitate accommodations that allow patients to participate and flourish in society. By the same logic, when approached by patients with a request for letters in support of reduced activity, you should consider not only the benefits but also the potential detriments of such disruptions. This may entail recommending temporary restrictions or modifications, as appropriate.<br/><br/><span class="bbody">Think beyond the patient</span> <br/><br/>Letter writing, particularly when recommending accommodations, can have implications beyond the patient.<sup>16</sup> Such letters may cause unintended societal harm. For example, others may have to assume additional responsibilities; competitive goods (eg, housing) may be rendered to the patient rather than to a person with greater needs; and workplace safety could be compromised due to absence. Consider not only the individual patient but also possible public health and societal effects of letter writing.<br/><br/><span class="bbody"><br/><br/>Deciding not to write</span>From an ethical perspective, a physician cannot be compelled to write a letter if such an undertaking violates a stronger moral obligation. An example of this is if writing a letter could cause significant harm to the patient or society, or if writing a letter might compromise a physician’s professionalism.<sup>19</sup> When you elect to not write a letter, the ethical principles of autonomy and truth telling dictate that you must inform your patients of this choice.<sup>6</sup> You should also provide an explanation to the patient as long as such information would not cause undue psychological or physical harm.<sup>20,21<br/><br/></sup><span class="bbody">Schedule time to write letters</span>Some physicians implement policies that all letters are to be completed during scheduled appointments. Others designate administrative time to complete requested letters. Finally, some physicians flexibly complete such requests between appointments or during other undedicated time slots. Any of these approaches are justifiable, though some urgent requests may require more immediate attention outside of appointments. Some physicians may choose to bill for the letter writing if completed outside an appointment and the patient is treated in private practice. Whatever your policy, inform patients of it at the beginning of care and remind them when appropriate, such as before completing a letter that may be billed.<br/><br/><span class="bbody"><br/><br/>Manage uncertainty</span>Always strive for objectivity in letter writing. However, some requests inherently hinge on subjective reports and assessments. For example, a patient may request an excuse letter due to feeling unwell. In the absence of objective findings, what should you do? We advise the following.</p> <p><span class="bbody">Acquire collateral information. </span>Adequate information is essential when making any medical recommendation. The same is true for writing letters. With the patient’s permission, you may need to contact relevant parties to better understand the circumstance or activity about which you are being asked to write a letter. For example, a patient may request leave from work due to injury. If the specific parameters of the work impeded by the injury are unclear to you, refrain from writing the letter and explain the rationale to the patient.<br/><br/><span class="bbody">Integrate prior knowledge of the patient.</span> No letter writing request exists in a vacuum. If you know the patient, the letter should be contextualized within the patient’s prior behaviors.</p> <p><span class="bbody">Stay within your scope</span>Given the various dilemmas and challenges, you may want to consider whether some letter writing is out of your professional scope.<sup>14-16</sup> One solution would be to leave such requests to other entities (eg, requiring employers to retain medical personnel with specialized skills in occupational evaluations) and make such recommendations to patients. Regardless, physicians should think carefully about their professional boundaries and scope regarding letter requests and adopt and implement a consistent standard for all patients.</p> <p>Regarding the letter requested by Ms. M, you should consider whether the appeal is consistent with the patient’s psychiatric illness. You should also consider whether you have sufficient knowledge about the patient’s living environment to support their claim. Such a letter should be written only if you understand both considerations. Regardless of your decision, you should explain your rationale to the patient.</p> <p> <strong>References</strong> </p> <p class="reference"> 1. West S, Friedman SH. To be or not to be: treating psychiatrist and expert witness. Psychiatric Times. 2007;24(6). Accessed March 14, 2023. https://www.psychiatrictimes.com/view/be-or-not-be-treating-psychiatrist-and-expert-witness<br/><br/> 2. Knoepflmacher D. ‘Medical necessity’ in psychiatry: whose definition is it anyway? Psychiatric News. 2016;51(18):12-14. https://psychnews.psychiatryonline.org/doi/10.1176/appi.pn.2016.9b14<br/><br/> 3. Lampe JR. Recent developments in marijuana law (LSB10859). Congressional Research Service. 2022. Accessed October 25, 2023. https://crsreports.congress.gov/product/pdf/LSB/LSB10859/2<br/><br/> 4. Brunnauer A, Buschert V, Segmiller F, et al. Mobility behaviour and driving status of patients with mental disorders – an exploratory study. Int J Psychiatry Clin Pract. 2016;20(1):40-46. doi:10.3109/13651501.2015.1089293<br/><br/> 5. Chiu CW, Law CK, Cheng AS. Driver assessment service for people with mental illness. Hong Kong J Occup Ther. 2019;32(2):77-83. doi:10.1177/1569186119886773<br/><br/> 6. Joshi KG. Service animals and emotional support animals: should you write that letter? Current Psychiatry. 2021;20(11):16-19. doi:10.12788/cp.0183<br/><br/> 7. <span class="iref">Tarasoff v Regents of University of California</span>, 17 Cal 3d 425, 551 P2d 334, 131 Cal. Rptr. 14 (Cal 1976).<br/><br/> 8. Black HC. Liability. <span class="iref">Black’s Law Dictionary</span>. Revised 4th ed. West Publishing; 1975:1060. <br/><br/> 9. American Academy of Psychiatry and the Law. Ethics guidelines for the practice of forensic psychiatry. 2005. Accessed March 15, 2023. https://www.aapl.org/ethics.htm<br/><br/> 10. Gold LH, Davidson JE. Do you understand your risk? Liability and third-party evaluations in civil litigation. J Am Acad Psychiatry Law. 2007;35(2):200-210.<br/><br/> 11. Schouten R. Approach to the patient seeking disability benefits. In: Stern TA, Herman JB, Slavin PL, eds. <span class="iref">The MGH Guide to Psychiatry in Primary Care</span>. McGraw Hill; 1998:121-126. <br/><br/> 12. Appelbaum PS. Law and psychiatry: liability for forensic evaluations: a word of caution. Psychiatr Serv. 2001;52(7):885-886. doi:10.1176/appi.ps.52.7.885<br/><br/> 13. Varkey B. Principles of clinical ethics and their application to practice. Med Princ Pract. 2021;30(1):17-28. doi:10.1159/000509119<br/><br/> 14. Mayhew HE, Nordlund DJ. Absenteeism certification: the physician’s role. J Fam Pract. 1988;26(6):651-655.<br/><br/> 15. Younggren JN, Boisvert JA, Boness CL. Examining emotional support animals and role conflicts in professional psychology. Prof Psychol Res Pr. 2016;47(4):255-260. doi:10.1037/pro0000083<br/><br/> 16. Carroll JD, Mohlenhoff BS, Kersten CM, et al. Laws and ethics related to emotional support animals. J Am Acad Psychiatry Law. 2020;48(4):509-518. doi:1-.29158/JAAPL.200047-20<br/><br/> 17. Strully KW. Job loss and health in the U.S. labor market. Demography. 2009;46(2):221-246. doi:10.1353/dem.0.0050<br/><br/> 18. Jurisic M, Bean M, Harbaugh J, et al. The personal physician’s role in helping patients with medical conditions stay at work or return to work. J Occup Environ Med. 2017;59(6):e125-131. doi:10.1097/JOM.0000000000001055<br/><br/> 19. Munyaradzi M. Critical reflections on the principle of beneficence in biomedicine. Pan Afr Med J. 2012;11:29. <br/><br/> 20. Beauchamp TL, Childress JF. <span class="iref">Principles of Biomedical Ethics</span>. 7th ed. Oxford University Press; 2012. <br/><br/> 21. Gold M. Is honesty always the best policy? Ethical aspects of truth telling. Intern Med J. 2004;34(9-10):578-580. doi:10.1111/j.1445-5994.2004.00673.x</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002653.SIG
Disable zoom
Off

Cannabis and schizophrenia: A complex relationship

Article Type
Changed
Wed, 12/06/2023 - 10:06
Display Headline
Cannabis and schizophrenia: A complex relationship

Approximately 1 in 200 individuals will be diagnosed with schizophrenia in their lifetime.1 DSM-5 criteria for the diagnosis of schizophrenia require the presence of ≥2 of 5 symptoms: delusions, hallucinations, disordered speech, grossly disorganized (or catatonic) behavior, and negative symptoms such as flat affect or avolition.2 Multiple studies have found increased rates of cannabis use among patients with schizophrenia. Because cognitive deficits are the chief predictor of clinical outcomes and quality of life in individuals with schizophrenia, the cognitive effects of cannabis use among these patients are of clinical significance.3 As legislation increasingly allows for the sale, possession, and consumption of cannabis, it is crucial to provide clinicians with evidence-based recommendations for treating patients who regularly use cannabis (approximately 8% of the adult population3). In this article, we analyze several peer-reviewed studies to investigate the impact of cannabis use on the onset and development of schizophrenia.

A look at substance-induced psychosis

Schizophrenia is associated with several structural brain changes, and some of these changes may be influenced by cannabis use (Box4). The biochemical etiology of schizophrenia is poorly understood but thought to involve dopamine, glutamate, serotonin, and gamma-aminobutyric acid. Certain positive symptoms, such as hallucinations, are uniquely human and difficult to study in animal models.5 Psychoactive substance use, especially cannabis, is frequently comorbid with schizophrenia. Additionally, certain individuals may be more predisposed to substance-induced psychosis than others based on genetic variation and underlying brain structure changes.4 Substance-induced psychosis is a psychotic state following the ingestion of a psychoactive substance or drug withdrawal lasting ≥48 hours.6 The psychoactive effects of cannabis have been associated with an exacerbation of existing schizophrenia symptoms.7 In 1998, Hall7 proposed 2 hypotheses to explain the relationship between cannabis and psychosis. The first was that heavy consumption of cannabis triggers a specific type of cannabis psychosis.7 The second was that cannabis use exacerbates existing schizophrenia, making the symptoms worse.7 Hall7 concluded that there was a complicated interaction among an individual’s vulnerability to their stressors, environment, and genetics.

Box

Schizophrenia, cannabis, and brain changes

Schizophrenia is associated with several structural changes in the brain, including lateral ventriculomegaly, reduced prefrontal cortex volume, and generalized atrophy. These changes may precede illness and act as a risk marker.4 A multivariate regression analysis that compared patients with schizophrenia who were cannabis users vs patients with schizophrenia who were nonusers found that those with high-level cannabis use had relatively higher left and right lateral ventricle volume (r = 0.208, P = .13, and r = 0.226, P = .007, respectively) as well as increased third ventricle volume (r = 0.271, P = .001).4 These changes were dose-dependent and may lead to worse disease outcomes.4

Cannabis, COMT, and homocysteine

Great advances have been made in our ability to examine the association between genetics and metabolism. One example of this is the interaction between the catechol-O-methyltransferase (COMT) gene and the active component of cannabis, delta-9-tetra­hydrocannabinol (THC). COMT codes for an enzyme that degrades cortical dopamine. The Val158Met polymorphism of this gene increases COMT activity, leading to increased dopamine catabolism, and thus decreased levels of extracellular dopamine, which induces an increase in mesolimbic dopaminergic activity, thereby increasing susceptibility to psychosis.3

 

In a study that genotyped 135 patients with schizophrenia, the Val158Met polymorphism was present in 29.63% of participants.3 Because THC can induce episodes of psychosis, individuals with this polymorphism may be at a higher risk of developing schizophrenia. Compared to Met carrier control participants with similar histories of cannabis consumption, those with the Val158Met polymorphism demonstrated markedly worse performance on tests of verbal fluency and processing speed.3 This is clinically significant because cognitive impairments are a major prognostic factor in schizophrenia, and identifying patients with this polymorphism could help personalize interventions for those who consume cannabis and are at risk of developing schizophrenia.

A study that evaluated 56 patients with first-episode schizophrenia found that having a history of cannabis abuse was associated with significantly higher levels of homocysteine as well as lower levels of high-density lipoprotein and vitamin B12.8 Homocysteine is an agonist at the glutamate binding site and a partial antagonist at the glycine co-agonist site in the N-methyl-D-aspartate receptor, which suggests that homocysteine may contribute to hypofunctioning of glutamate transmission; this is implicated in the development of schizophrenia. These increases in homocysteine are also found in siblings of patients with schizophrenia, which indicates a possible association between the methylenetetrahydrofolate (MTHFR) gene and schizophrenia.8

The C677T polymorphism in MTHFR may predict the risk of developing metabolic syndrome in patients taking second-generation antipsychotics.8 Elevations in homocysteine by as little as 5 μmol/L may increase schizophrenia risk by 70% compared to controls, possibly due to homocysteine initiating neuronal apoptosis, catalyzing dysfunction of the mitochondria, or increasing oxidative stress.8 There is a positive correlation between homocysteine levels and severity of negative symptoms (P = .006) and general psychopathology (P = .008) of schizophrenia when analyzed using the Positive and Negative Syndrome Scale.8 Negative symptoms such as blunted affect, apathy, anhedonia, and loss of motivation significantly impact the social and economic outcomes of patients diagnosed with schizophrenia.

Research paints a mixed picture

A Danish study analyzed the rates of conversion to schizophrenia or bipolar disorder (BD) among 6,788 individuals who received a diagnosis of substance-induced psychosis from 1994 to 2014.6 Ten comparison participants were selected for each case participant, matched on sex and year/month of birth. Participants were followed until the first occurrence of schizophrenia or BD, death, or emigration from Denmark. Substances implicated in the initial psychotic episode included cannabis, alcohol, opioids, sedatives, cocaine, amphetamines, hallucinogens, and combinations of substances.

Continue to: The overall conversion rate...

 

 

The overall conversion rate over 20 years was 32.2% (95% CI, 29.7 to 34.9), with 26.0% developing schizophrenia vs 8.4% developing BD.6 Of the substances involved, cannabis was the most common, implicated in 41.2% (95% CI, 36.6 to 46.2) of cases.6 One-half of male patients converted within 2.0 years and one-half of female patients converted within 4.4 years after a cannabis-induced psychosis.6

This study had several limitations. It could not account for any short-term psychotic symptoms experienced by the general population, especially after cannabis use. Such patients might not seek treatment. Thus, the results might not be generalizable to the general population. The study did not evaluate if conversion rates differed based on continued substance use following the psychosis episode, or the amount of each substance taken prior to the episode. Dose-dependence was not well elucidated, and this study only looked at patients from Denmark and did not account for socioeconomic status.6

Another Danish study looked at the influences of gender and cannabis use in the early course of the disease in 133 patients with schizophrenia.9 These researchers found that male gender was a significant predictor of earlier onset of dysfunction socially and in the workplace, as well as a higher risk of developing negative symptoms. However, compared to gender, cannabis use was a stronger predictor of age at first psychotic episode. For cannabis users, the median age of onset of negative symptoms was 23.7, compared to 38.4 for nonusers (P < .001).9

Cannabis use is significantly elevated among individuals with psychosis, with a 12-month prevalence of 29.2% compared to 4.0% among the general population of the United States.10 In a study that assessed 229 patients with a schizophrenia spectrum disorder during their first hospitalization and 6 months, 2 years, 4 years, and 10 years later, Foti et al10 found that the lifetime rate of cannabis use was 66.2%. Survival analysis found cannabis use doubled the risk of the onset of psychosis compared to nonusers of the same age (hazard ratio [HR] = 1.97; 95% CI, 1.48 to 2.62, P < .001), even after adjusting for socioeconomic status, age, and gender (HR = 1.34; 95% CI, 1.01 to 1.77, P < .05).10 Additionally, Foti et al10 found significant positive correlations between psychotic symptoms and cannabis use in patients with schizophrenia over the course of 10 years. An increase in symptoms was associated with a higher likelihood of cannabis use, and a decrease in symptoms was correlated with a lower likelihood of use (adjusted odds ratio = 1.64; 95% CI, 1.12 to 2.43, P < .0125).10

Ortiz-Medina et al11 conducted a meta-analysis of 22 studies of 15 cohorts from healthy populations and 12 other cohort follow-up studies that evaluated the onset of psychotic symptoms in individuals who used cannabis. Most studies found associations between cannabis use and the onset of symptoms of schizophrenia, and most determined cannabis was also a major risk factor for other psychotic disorders. Analyses of dose-dependence indicated that repeated cannabis use increased the risk of developing psychotic symptoms. This risk is increased when an individual starts using cannabis before age 15.11 Age seemed to be a stronger predictor of onset and outcome than sex, with no significant differences between men and women. One study in this review found that approximately 8% to 13% cases of schizophrenia may have been solely due to cannabis.11 The most significant limitation to the studies analyzed in this review is that retrospective studies utilize self-reported questionnaires.

Continue to: Other researchers have found...

 

 

Other researchers have found it would take a relatively high number of individuals to stop using cannabis to prevent 1 case of schizophrenia. In a study of data from England and Wales, Hickman et al12 evaluated the best available estimates of the incidence of schizophrenia, rates of heavy and light cannabis use, and risk that cannabis causes schizophrenia to determine the number needed to prevent (NNP) 1 case of schizophrenia. They estimated that it would require approximately 2,800 men age 20 to 24 (90% CI, 2,018 to 4,530) and 4,700 men age 35 to 39 (90% CI, 3,114 to 8,416) who heavily used cannabis to stop their consumption to prevent 1 case of schizophrenia.12 For women with heavy cannabis use, the mean NNP was 5,470 for women age 25 to 29 (90% CI, 3,640 to 9,839) and 10,870 for women age 35 to 39 (90% CI, 6,786 to 22,732).12 For light cannabis users, the NNP was 4 to 5 times higher than the NNP for heavy cannabis users. This suggests that clinical interventions aimed at preventing dependence on cannabis would be more effective than interventions aimed at eliminating cannabis use.

Medical cannabis and increased potency

In recent years, the use of medical cannabis, which is used to address adverse effects of chemotherapy as well as neuropathic pain, Parkinson’s disease, and epilepsy, has been increasing.13 However, there is a lack of well-conducted randomized clinical trials evaluating medical cannabis’ efficacy and safety. As medical cannabis continues to gain public acceptance and more states permit its legal use, patients and physicians should be fully informed of the known adverse effects, including impaired attention, learning, and motivation.13

Several studies have drawn attention to the dose-dependence of many of cannabis’ effects. Since at least the 1960s, the concentration of THC in cannabis has increased substantially, thus increasing its potency. Based on 66,747 samples across 8 studies, 1 meta-analysis estimated that THC concentrations in herbal cannabis increased by 0.29% (P < .001) each year between 1970 and 2017.14 Similarly, THC concentrations in cannabis resins were found to have increased by 0.57% (P = .017) each year between 1975 and 2017.14 Cannabis products with high concentrations of THC carry an increased risk of addiction and mental health disorders.14

 

Identifying those at highest risk

Despite ongoing research, scientific consensus on the relationship of cannabis to schizophrenia and psychosis has yet to be reached. The disparity between the relatively high prevalence of regular adult use of cannabis (8%7)and the low incidence of cannabis-induced psychosis suggests that cannabis use alone is unlikely to lead to episodes of psychosis in individuals who are not predisposed to such episodes. Sarrazin et al15 evaluated 170 patients with schizophrenia, 31 of whom had cannabis use disorder. They found no significant difference in lifetime symptom dimensions between groups, and proposed that cannabis-associated schizophrenia should not be categorized as a distinct clinical entity of schizophrenia with specific features.15

Policies that encourage follow-up of patients after episodes of drug-induced psychosis may mitigate the adverse social and economic effects of schizophrenia. Currently, these policies are not widely implemented in health care institutions, possibly because psychotic symptoms may fade after the drug’s effects have dissipated. Despite this, these patients are at high risk of developing schizophrenia and self-harm. New-onset schizophrenia should be promptly identified because delayed diagnosis is associated with worse prognosis.6 Additionally, identifying genetic susceptibilities to schizophrenia—such as the Val158Met polymorphisms—in individuals who use cannabis could help clinicians manage or slow the onset or progression of schizophrenia.3 Motivational interviewing strategies should be used to minimize or eliminate cannabis use in individuals with active schizophrenia or psychosis, thus preventing worse outcomes.

Bottom Line

Identifying susceptibilities to schizophrenia may guide interventions in patients who use cannabis. Several large studies have suggested that cannabis use may exacerbate symptoms and worsen the prognosis of schizophrenia. Motivational interviewing strategies aimed at minimizing cannabis use may improve outcomes in patients with schizophrenia.

Related Resources

  • Khokhar JY, Dwiel LL, Henricks AM, et al. The link between schizophrenia and substance use disorder: a unifying hypothesis. Schizophr Res. 2018;194:78-85. doi:10.1016/j. schres.2017.04.016
  • Otite ES, Solanky A, Doumas S. Adolescents, THC, and the risk of psychosis. Current Psychiatry. 2021;20(12):e1-e2. doi:10.12788/cp.0197

[embed:render:related:node:249151]

References

1. Simeone JC, Ward AJ, Rotella P, et al. An evaluation of variation in published estimates of schizophrenia prevalence from 1990-2013: a systematic literature review. BMC Psychiatry. 2015;15(1):193. doi:10.1186/s12888-015-0578-7

2. Tandon R, Gaebel W, Barch DM, et al. Definition and description of schizophrenia in the DSM-5. Schizophr Res. 2013;150(1):3-10. doi:10.1016/j.schres.2013.05.028

3. Bosia M, Buonocore M, Bechi M, et al. Schizophrenia, cannabis use and catechol-O-methyltransferase (COMT): modeling the interplay on cognition. Prog Neuropsychopharmacol Biol Psychiatry. 2019;92:363-368. doi:10.1016/j.pnpbp.2019.02.009

4. Welch KA, McIntosh AM, Job DE, et al. The impact of substance use on brain structure in people at high risk of developing schizophrenia. Schizophr Bull. 2011;37(5):1066-1076. doi:10.1093/schbul/sbq013

5. Winship IR, Dursun SM, Baker GB, et al. An overview of animal models related to schizophrenia. Can J Psychiatry. 2019;64(1):5-17. doi:10.1177/0706743718773728

6. Starzer MSK, Nordentoft M, Hjorthøj C. Rates and predictors of conversion to schizophrenia or bipolar disorder following substance-induced psychosis. Am J Psychiatry. 2018;175(4):343-350. doi:10.1176/appi.ajp.2017.17020223

7. Hall W. Cannabis use and psychosis. Drug Alcohol Rev. 1998;17(4):433-444. doi:10.1080/09595239800187271

8. Misiak B, Frydecka D, Slezak R, et al. Elevated homocysteine level in first-episode schizophrenia patients—the relevance of family history of schizophrenia and lifetime diagnosis of cannabis abuse. Metab Brain Dis. 2014;29(3):661-670. doi:10.1007/s11011-014-9534-3

9. Veen ND, Selten J, van der Tweel I, et al. Cannabis use and age at onset of schizophrenia. Am J Psychiatry. 2004;161(3):501-506. doi:10.1176/appi.ajp.161.3.501

10. Foti DJ, Kotov R, Guey LT, et al. Cannabis use and the course of schizophrenia: 10-year follow-up after first hospitalization. Am J Psychiatry. 2010;167(8):987-993. doi:10.1176/appi.ajp.2010.09020189

11. Ortiz-Medina MB, Perea M, Torales J, et al. Cannabis consumption and psychosis or schizophrenia development. Int J Soc Psychiatry. 2018;64(7):690-704. doi:10.1177/0020764018801690

12. Hickman M, Vickerman P, Macleod J, et al. If cannabis caused schizophrenia—how many cannabis users may need to be prevented in order to prevent one case of schizophrenia? England and Wales calculations. Addiction. 2009;104(11):1856-1861. doi:10.1111/j.1360-0443.2009.02736.x

13. Gupta S, Phalen T, Gupta S. Medical marijuana: do the benefits outweigh the risks? Current Psychiatry. 2018;17(1):34-41.

14. Freeman TP, Craft S, Wilson J, et al. Changes in delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) concentrations in cannabis over time: systematic review and meta-analysis. Addiction. 2021;116(5):1000-1010. doi:10.1111/add.15253

15. Sarrazin S, Louppe F, Doukhan R, et al. A clinical comparison of schizophrenia with and without pre-onset cannabis use disorder: a retrospective cohort study using categorical and dimensional approaches. Ann Gen Psychiatry. 2015;14:44. doi:10.1186/s12991-015-0083-x

Article PDF
Author and Disclosure Information

Joseph M. Maes, BS
Medical Student
Creighton University School of Medicine
Omaha, Nebraska

Charlotte A. Ritchie, MD
PGY-1 Psychiatry Resident
Department of Psychiatry
University of Nebraska Medical Center
Omaha, Nebraska

Sriram Ramaswamy, MD
Professor of Psychiatry
Department of Psychiatry
Creighton University School of Medicine
Omaha, Nebraska

Disclosures
Dr. Ramaswamy receives research support from Bristol Myers Squibb and Forest Pharmaceuticals. Joseph M. Maes and Dr. Ritchie report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Issue
Current Psychiatry - 22(12)
Publications
Topics
Page Number
43-47
Sections
Author and Disclosure Information

Joseph M. Maes, BS
Medical Student
Creighton University School of Medicine
Omaha, Nebraska

Charlotte A. Ritchie, MD
PGY-1 Psychiatry Resident
Department of Psychiatry
University of Nebraska Medical Center
Omaha, Nebraska

Sriram Ramaswamy, MD
Professor of Psychiatry
Department of Psychiatry
Creighton University School of Medicine
Omaha, Nebraska

Disclosures
Dr. Ramaswamy receives research support from Bristol Myers Squibb and Forest Pharmaceuticals. Joseph M. Maes and Dr. Ritchie report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Author and Disclosure Information

Joseph M. Maes, BS
Medical Student
Creighton University School of Medicine
Omaha, Nebraska

Charlotte A. Ritchie, MD
PGY-1 Psychiatry Resident
Department of Psychiatry
University of Nebraska Medical Center
Omaha, Nebraska

Sriram Ramaswamy, MD
Professor of Psychiatry
Department of Psychiatry
Creighton University School of Medicine
Omaha, Nebraska

Disclosures
Dr. Ramaswamy receives research support from Bristol Myers Squibb and Forest Pharmaceuticals. Joseph M. Maes and Dr. Ritchie report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Article PDF
Article PDF

Approximately 1 in 200 individuals will be diagnosed with schizophrenia in their lifetime.1 DSM-5 criteria for the diagnosis of schizophrenia require the presence of ≥2 of 5 symptoms: delusions, hallucinations, disordered speech, grossly disorganized (or catatonic) behavior, and negative symptoms such as flat affect or avolition.2 Multiple studies have found increased rates of cannabis use among patients with schizophrenia. Because cognitive deficits are the chief predictor of clinical outcomes and quality of life in individuals with schizophrenia, the cognitive effects of cannabis use among these patients are of clinical significance.3 As legislation increasingly allows for the sale, possession, and consumption of cannabis, it is crucial to provide clinicians with evidence-based recommendations for treating patients who regularly use cannabis (approximately 8% of the adult population3). In this article, we analyze several peer-reviewed studies to investigate the impact of cannabis use on the onset and development of schizophrenia.

A look at substance-induced psychosis

Schizophrenia is associated with several structural brain changes, and some of these changes may be influenced by cannabis use (Box4). The biochemical etiology of schizophrenia is poorly understood but thought to involve dopamine, glutamate, serotonin, and gamma-aminobutyric acid. Certain positive symptoms, such as hallucinations, are uniquely human and difficult to study in animal models.5 Psychoactive substance use, especially cannabis, is frequently comorbid with schizophrenia. Additionally, certain individuals may be more predisposed to substance-induced psychosis than others based on genetic variation and underlying brain structure changes.4 Substance-induced psychosis is a psychotic state following the ingestion of a psychoactive substance or drug withdrawal lasting ≥48 hours.6 The psychoactive effects of cannabis have been associated with an exacerbation of existing schizophrenia symptoms.7 In 1998, Hall7 proposed 2 hypotheses to explain the relationship between cannabis and psychosis. The first was that heavy consumption of cannabis triggers a specific type of cannabis psychosis.7 The second was that cannabis use exacerbates existing schizophrenia, making the symptoms worse.7 Hall7 concluded that there was a complicated interaction among an individual’s vulnerability to their stressors, environment, and genetics.

Box

Schizophrenia, cannabis, and brain changes

Schizophrenia is associated with several structural changes in the brain, including lateral ventriculomegaly, reduced prefrontal cortex volume, and generalized atrophy. These changes may precede illness and act as a risk marker.4 A multivariate regression analysis that compared patients with schizophrenia who were cannabis users vs patients with schizophrenia who were nonusers found that those with high-level cannabis use had relatively higher left and right lateral ventricle volume (r = 0.208, P = .13, and r = 0.226, P = .007, respectively) as well as increased third ventricle volume (r = 0.271, P = .001).4 These changes were dose-dependent and may lead to worse disease outcomes.4

Cannabis, COMT, and homocysteine

Great advances have been made in our ability to examine the association between genetics and metabolism. One example of this is the interaction between the catechol-O-methyltransferase (COMT) gene and the active component of cannabis, delta-9-tetra­hydrocannabinol (THC). COMT codes for an enzyme that degrades cortical dopamine. The Val158Met polymorphism of this gene increases COMT activity, leading to increased dopamine catabolism, and thus decreased levels of extracellular dopamine, which induces an increase in mesolimbic dopaminergic activity, thereby increasing susceptibility to psychosis.3

 

In a study that genotyped 135 patients with schizophrenia, the Val158Met polymorphism was present in 29.63% of participants.3 Because THC can induce episodes of psychosis, individuals with this polymorphism may be at a higher risk of developing schizophrenia. Compared to Met carrier control participants with similar histories of cannabis consumption, those with the Val158Met polymorphism demonstrated markedly worse performance on tests of verbal fluency and processing speed.3 This is clinically significant because cognitive impairments are a major prognostic factor in schizophrenia, and identifying patients with this polymorphism could help personalize interventions for those who consume cannabis and are at risk of developing schizophrenia.

A study that evaluated 56 patients with first-episode schizophrenia found that having a history of cannabis abuse was associated with significantly higher levels of homocysteine as well as lower levels of high-density lipoprotein and vitamin B12.8 Homocysteine is an agonist at the glutamate binding site and a partial antagonist at the glycine co-agonist site in the N-methyl-D-aspartate receptor, which suggests that homocysteine may contribute to hypofunctioning of glutamate transmission; this is implicated in the development of schizophrenia. These increases in homocysteine are also found in siblings of patients with schizophrenia, which indicates a possible association between the methylenetetrahydrofolate (MTHFR) gene and schizophrenia.8

The C677T polymorphism in MTHFR may predict the risk of developing metabolic syndrome in patients taking second-generation antipsychotics.8 Elevations in homocysteine by as little as 5 μmol/L may increase schizophrenia risk by 70% compared to controls, possibly due to homocysteine initiating neuronal apoptosis, catalyzing dysfunction of the mitochondria, or increasing oxidative stress.8 There is a positive correlation between homocysteine levels and severity of negative symptoms (P = .006) and general psychopathology (P = .008) of schizophrenia when analyzed using the Positive and Negative Syndrome Scale.8 Negative symptoms such as blunted affect, apathy, anhedonia, and loss of motivation significantly impact the social and economic outcomes of patients diagnosed with schizophrenia.

Research paints a mixed picture

A Danish study analyzed the rates of conversion to schizophrenia or bipolar disorder (BD) among 6,788 individuals who received a diagnosis of substance-induced psychosis from 1994 to 2014.6 Ten comparison participants were selected for each case participant, matched on sex and year/month of birth. Participants were followed until the first occurrence of schizophrenia or BD, death, or emigration from Denmark. Substances implicated in the initial psychotic episode included cannabis, alcohol, opioids, sedatives, cocaine, amphetamines, hallucinogens, and combinations of substances.

Continue to: The overall conversion rate...

 

 

The overall conversion rate over 20 years was 32.2% (95% CI, 29.7 to 34.9), with 26.0% developing schizophrenia vs 8.4% developing BD.6 Of the substances involved, cannabis was the most common, implicated in 41.2% (95% CI, 36.6 to 46.2) of cases.6 One-half of male patients converted within 2.0 years and one-half of female patients converted within 4.4 years after a cannabis-induced psychosis.6

This study had several limitations. It could not account for any short-term psychotic symptoms experienced by the general population, especially after cannabis use. Such patients might not seek treatment. Thus, the results might not be generalizable to the general population. The study did not evaluate if conversion rates differed based on continued substance use following the psychosis episode, or the amount of each substance taken prior to the episode. Dose-dependence was not well elucidated, and this study only looked at patients from Denmark and did not account for socioeconomic status.6

Another Danish study looked at the influences of gender and cannabis use in the early course of the disease in 133 patients with schizophrenia.9 These researchers found that male gender was a significant predictor of earlier onset of dysfunction socially and in the workplace, as well as a higher risk of developing negative symptoms. However, compared to gender, cannabis use was a stronger predictor of age at first psychotic episode. For cannabis users, the median age of onset of negative symptoms was 23.7, compared to 38.4 for nonusers (P < .001).9

Cannabis use is significantly elevated among individuals with psychosis, with a 12-month prevalence of 29.2% compared to 4.0% among the general population of the United States.10 In a study that assessed 229 patients with a schizophrenia spectrum disorder during their first hospitalization and 6 months, 2 years, 4 years, and 10 years later, Foti et al10 found that the lifetime rate of cannabis use was 66.2%. Survival analysis found cannabis use doubled the risk of the onset of psychosis compared to nonusers of the same age (hazard ratio [HR] = 1.97; 95% CI, 1.48 to 2.62, P < .001), even after adjusting for socioeconomic status, age, and gender (HR = 1.34; 95% CI, 1.01 to 1.77, P < .05).10 Additionally, Foti et al10 found significant positive correlations between psychotic symptoms and cannabis use in patients with schizophrenia over the course of 10 years. An increase in symptoms was associated with a higher likelihood of cannabis use, and a decrease in symptoms was correlated with a lower likelihood of use (adjusted odds ratio = 1.64; 95% CI, 1.12 to 2.43, P < .0125).10

Ortiz-Medina et al11 conducted a meta-analysis of 22 studies of 15 cohorts from healthy populations and 12 other cohort follow-up studies that evaluated the onset of psychotic symptoms in individuals who used cannabis. Most studies found associations between cannabis use and the onset of symptoms of schizophrenia, and most determined cannabis was also a major risk factor for other psychotic disorders. Analyses of dose-dependence indicated that repeated cannabis use increased the risk of developing psychotic symptoms. This risk is increased when an individual starts using cannabis before age 15.11 Age seemed to be a stronger predictor of onset and outcome than sex, with no significant differences between men and women. One study in this review found that approximately 8% to 13% cases of schizophrenia may have been solely due to cannabis.11 The most significant limitation to the studies analyzed in this review is that retrospective studies utilize self-reported questionnaires.

Continue to: Other researchers have found...

 

 

Other researchers have found it would take a relatively high number of individuals to stop using cannabis to prevent 1 case of schizophrenia. In a study of data from England and Wales, Hickman et al12 evaluated the best available estimates of the incidence of schizophrenia, rates of heavy and light cannabis use, and risk that cannabis causes schizophrenia to determine the number needed to prevent (NNP) 1 case of schizophrenia. They estimated that it would require approximately 2,800 men age 20 to 24 (90% CI, 2,018 to 4,530) and 4,700 men age 35 to 39 (90% CI, 3,114 to 8,416) who heavily used cannabis to stop their consumption to prevent 1 case of schizophrenia.12 For women with heavy cannabis use, the mean NNP was 5,470 for women age 25 to 29 (90% CI, 3,640 to 9,839) and 10,870 for women age 35 to 39 (90% CI, 6,786 to 22,732).12 For light cannabis users, the NNP was 4 to 5 times higher than the NNP for heavy cannabis users. This suggests that clinical interventions aimed at preventing dependence on cannabis would be more effective than interventions aimed at eliminating cannabis use.

Medical cannabis and increased potency

In recent years, the use of medical cannabis, which is used to address adverse effects of chemotherapy as well as neuropathic pain, Parkinson’s disease, and epilepsy, has been increasing.13 However, there is a lack of well-conducted randomized clinical trials evaluating medical cannabis’ efficacy and safety. As medical cannabis continues to gain public acceptance and more states permit its legal use, patients and physicians should be fully informed of the known adverse effects, including impaired attention, learning, and motivation.13

Several studies have drawn attention to the dose-dependence of many of cannabis’ effects. Since at least the 1960s, the concentration of THC in cannabis has increased substantially, thus increasing its potency. Based on 66,747 samples across 8 studies, 1 meta-analysis estimated that THC concentrations in herbal cannabis increased by 0.29% (P < .001) each year between 1970 and 2017.14 Similarly, THC concentrations in cannabis resins were found to have increased by 0.57% (P = .017) each year between 1975 and 2017.14 Cannabis products with high concentrations of THC carry an increased risk of addiction and mental health disorders.14

 

Identifying those at highest risk

Despite ongoing research, scientific consensus on the relationship of cannabis to schizophrenia and psychosis has yet to be reached. The disparity between the relatively high prevalence of regular adult use of cannabis (8%7)and the low incidence of cannabis-induced psychosis suggests that cannabis use alone is unlikely to lead to episodes of psychosis in individuals who are not predisposed to such episodes. Sarrazin et al15 evaluated 170 patients with schizophrenia, 31 of whom had cannabis use disorder. They found no significant difference in lifetime symptom dimensions between groups, and proposed that cannabis-associated schizophrenia should not be categorized as a distinct clinical entity of schizophrenia with specific features.15

Policies that encourage follow-up of patients after episodes of drug-induced psychosis may mitigate the adverse social and economic effects of schizophrenia. Currently, these policies are not widely implemented in health care institutions, possibly because psychotic symptoms may fade after the drug’s effects have dissipated. Despite this, these patients are at high risk of developing schizophrenia and self-harm. New-onset schizophrenia should be promptly identified because delayed diagnosis is associated with worse prognosis.6 Additionally, identifying genetic susceptibilities to schizophrenia—such as the Val158Met polymorphisms—in individuals who use cannabis could help clinicians manage or slow the onset or progression of schizophrenia.3 Motivational interviewing strategies should be used to minimize or eliminate cannabis use in individuals with active schizophrenia or psychosis, thus preventing worse outcomes.

Bottom Line

Identifying susceptibilities to schizophrenia may guide interventions in patients who use cannabis. Several large studies have suggested that cannabis use may exacerbate symptoms and worsen the prognosis of schizophrenia. Motivational interviewing strategies aimed at minimizing cannabis use may improve outcomes in patients with schizophrenia.

Related Resources

  • Khokhar JY, Dwiel LL, Henricks AM, et al. The link between schizophrenia and substance use disorder: a unifying hypothesis. Schizophr Res. 2018;194:78-85. doi:10.1016/j. schres.2017.04.016
  • Otite ES, Solanky A, Doumas S. Adolescents, THC, and the risk of psychosis. Current Psychiatry. 2021;20(12):e1-e2. doi:10.12788/cp.0197

[embed:render:related:node:249151]

Approximately 1 in 200 individuals will be diagnosed with schizophrenia in their lifetime.1 DSM-5 criteria for the diagnosis of schizophrenia require the presence of ≥2 of 5 symptoms: delusions, hallucinations, disordered speech, grossly disorganized (or catatonic) behavior, and negative symptoms such as flat affect or avolition.2 Multiple studies have found increased rates of cannabis use among patients with schizophrenia. Because cognitive deficits are the chief predictor of clinical outcomes and quality of life in individuals with schizophrenia, the cognitive effects of cannabis use among these patients are of clinical significance.3 As legislation increasingly allows for the sale, possession, and consumption of cannabis, it is crucial to provide clinicians with evidence-based recommendations for treating patients who regularly use cannabis (approximately 8% of the adult population3). In this article, we analyze several peer-reviewed studies to investigate the impact of cannabis use on the onset and development of schizophrenia.

A look at substance-induced psychosis

Schizophrenia is associated with several structural brain changes, and some of these changes may be influenced by cannabis use (Box4). The biochemical etiology of schizophrenia is poorly understood but thought to involve dopamine, glutamate, serotonin, and gamma-aminobutyric acid. Certain positive symptoms, such as hallucinations, are uniquely human and difficult to study in animal models.5 Psychoactive substance use, especially cannabis, is frequently comorbid with schizophrenia. Additionally, certain individuals may be more predisposed to substance-induced psychosis than others based on genetic variation and underlying brain structure changes.4 Substance-induced psychosis is a psychotic state following the ingestion of a psychoactive substance or drug withdrawal lasting ≥48 hours.6 The psychoactive effects of cannabis have been associated with an exacerbation of existing schizophrenia symptoms.7 In 1998, Hall7 proposed 2 hypotheses to explain the relationship between cannabis and psychosis. The first was that heavy consumption of cannabis triggers a specific type of cannabis psychosis.7 The second was that cannabis use exacerbates existing schizophrenia, making the symptoms worse.7 Hall7 concluded that there was a complicated interaction among an individual’s vulnerability to their stressors, environment, and genetics.

Box

Schizophrenia, cannabis, and brain changes

Schizophrenia is associated with several structural changes in the brain, including lateral ventriculomegaly, reduced prefrontal cortex volume, and generalized atrophy. These changes may precede illness and act as a risk marker.4 A multivariate regression analysis that compared patients with schizophrenia who were cannabis users vs patients with schizophrenia who were nonusers found that those with high-level cannabis use had relatively higher left and right lateral ventricle volume (r = 0.208, P = .13, and r = 0.226, P = .007, respectively) as well as increased third ventricle volume (r = 0.271, P = .001).4 These changes were dose-dependent and may lead to worse disease outcomes.4

Cannabis, COMT, and homocysteine

Great advances have been made in our ability to examine the association between genetics and metabolism. One example of this is the interaction between the catechol-O-methyltransferase (COMT) gene and the active component of cannabis, delta-9-tetra­hydrocannabinol (THC). COMT codes for an enzyme that degrades cortical dopamine. The Val158Met polymorphism of this gene increases COMT activity, leading to increased dopamine catabolism, and thus decreased levels of extracellular dopamine, which induces an increase in mesolimbic dopaminergic activity, thereby increasing susceptibility to psychosis.3

 

In a study that genotyped 135 patients with schizophrenia, the Val158Met polymorphism was present in 29.63% of participants.3 Because THC can induce episodes of psychosis, individuals with this polymorphism may be at a higher risk of developing schizophrenia. Compared to Met carrier control participants with similar histories of cannabis consumption, those with the Val158Met polymorphism demonstrated markedly worse performance on tests of verbal fluency and processing speed.3 This is clinically significant because cognitive impairments are a major prognostic factor in schizophrenia, and identifying patients with this polymorphism could help personalize interventions for those who consume cannabis and are at risk of developing schizophrenia.

A study that evaluated 56 patients with first-episode schizophrenia found that having a history of cannabis abuse was associated with significantly higher levels of homocysteine as well as lower levels of high-density lipoprotein and vitamin B12.8 Homocysteine is an agonist at the glutamate binding site and a partial antagonist at the glycine co-agonist site in the N-methyl-D-aspartate receptor, which suggests that homocysteine may contribute to hypofunctioning of glutamate transmission; this is implicated in the development of schizophrenia. These increases in homocysteine are also found in siblings of patients with schizophrenia, which indicates a possible association between the methylenetetrahydrofolate (MTHFR) gene and schizophrenia.8

The C677T polymorphism in MTHFR may predict the risk of developing metabolic syndrome in patients taking second-generation antipsychotics.8 Elevations in homocysteine by as little as 5 μmol/L may increase schizophrenia risk by 70% compared to controls, possibly due to homocysteine initiating neuronal apoptosis, catalyzing dysfunction of the mitochondria, or increasing oxidative stress.8 There is a positive correlation between homocysteine levels and severity of negative symptoms (P = .006) and general psychopathology (P = .008) of schizophrenia when analyzed using the Positive and Negative Syndrome Scale.8 Negative symptoms such as blunted affect, apathy, anhedonia, and loss of motivation significantly impact the social and economic outcomes of patients diagnosed with schizophrenia.

Research paints a mixed picture

A Danish study analyzed the rates of conversion to schizophrenia or bipolar disorder (BD) among 6,788 individuals who received a diagnosis of substance-induced psychosis from 1994 to 2014.6 Ten comparison participants were selected for each case participant, matched on sex and year/month of birth. Participants were followed until the first occurrence of schizophrenia or BD, death, or emigration from Denmark. Substances implicated in the initial psychotic episode included cannabis, alcohol, opioids, sedatives, cocaine, amphetamines, hallucinogens, and combinations of substances.

Continue to: The overall conversion rate...

 

 

The overall conversion rate over 20 years was 32.2% (95% CI, 29.7 to 34.9), with 26.0% developing schizophrenia vs 8.4% developing BD.6 Of the substances involved, cannabis was the most common, implicated in 41.2% (95% CI, 36.6 to 46.2) of cases.6 One-half of male patients converted within 2.0 years and one-half of female patients converted within 4.4 years after a cannabis-induced psychosis.6

This study had several limitations. It could not account for any short-term psychotic symptoms experienced by the general population, especially after cannabis use. Such patients might not seek treatment. Thus, the results might not be generalizable to the general population. The study did not evaluate if conversion rates differed based on continued substance use following the psychosis episode, or the amount of each substance taken prior to the episode. Dose-dependence was not well elucidated, and this study only looked at patients from Denmark and did not account for socioeconomic status.6

Another Danish study looked at the influences of gender and cannabis use in the early course of the disease in 133 patients with schizophrenia.9 These researchers found that male gender was a significant predictor of earlier onset of dysfunction socially and in the workplace, as well as a higher risk of developing negative symptoms. However, compared to gender, cannabis use was a stronger predictor of age at first psychotic episode. For cannabis users, the median age of onset of negative symptoms was 23.7, compared to 38.4 for nonusers (P < .001).9

Cannabis use is significantly elevated among individuals with psychosis, with a 12-month prevalence of 29.2% compared to 4.0% among the general population of the United States.10 In a study that assessed 229 patients with a schizophrenia spectrum disorder during their first hospitalization and 6 months, 2 years, 4 years, and 10 years later, Foti et al10 found that the lifetime rate of cannabis use was 66.2%. Survival analysis found cannabis use doubled the risk of the onset of psychosis compared to nonusers of the same age (hazard ratio [HR] = 1.97; 95% CI, 1.48 to 2.62, P < .001), even after adjusting for socioeconomic status, age, and gender (HR = 1.34; 95% CI, 1.01 to 1.77, P < .05).10 Additionally, Foti et al10 found significant positive correlations between psychotic symptoms and cannabis use in patients with schizophrenia over the course of 10 years. An increase in symptoms was associated with a higher likelihood of cannabis use, and a decrease in symptoms was correlated with a lower likelihood of use (adjusted odds ratio = 1.64; 95% CI, 1.12 to 2.43, P < .0125).10

Ortiz-Medina et al11 conducted a meta-analysis of 22 studies of 15 cohorts from healthy populations and 12 other cohort follow-up studies that evaluated the onset of psychotic symptoms in individuals who used cannabis. Most studies found associations between cannabis use and the onset of symptoms of schizophrenia, and most determined cannabis was also a major risk factor for other psychotic disorders. Analyses of dose-dependence indicated that repeated cannabis use increased the risk of developing psychotic symptoms. This risk is increased when an individual starts using cannabis before age 15.11 Age seemed to be a stronger predictor of onset and outcome than sex, with no significant differences between men and women. One study in this review found that approximately 8% to 13% cases of schizophrenia may have been solely due to cannabis.11 The most significant limitation to the studies analyzed in this review is that retrospective studies utilize self-reported questionnaires.

Continue to: Other researchers have found...

 

 

Other researchers have found it would take a relatively high number of individuals to stop using cannabis to prevent 1 case of schizophrenia. In a study of data from England and Wales, Hickman et al12 evaluated the best available estimates of the incidence of schizophrenia, rates of heavy and light cannabis use, and risk that cannabis causes schizophrenia to determine the number needed to prevent (NNP) 1 case of schizophrenia. They estimated that it would require approximately 2,800 men age 20 to 24 (90% CI, 2,018 to 4,530) and 4,700 men age 35 to 39 (90% CI, 3,114 to 8,416) who heavily used cannabis to stop their consumption to prevent 1 case of schizophrenia.12 For women with heavy cannabis use, the mean NNP was 5,470 for women age 25 to 29 (90% CI, 3,640 to 9,839) and 10,870 for women age 35 to 39 (90% CI, 6,786 to 22,732).12 For light cannabis users, the NNP was 4 to 5 times higher than the NNP for heavy cannabis users. This suggests that clinical interventions aimed at preventing dependence on cannabis would be more effective than interventions aimed at eliminating cannabis use.

Medical cannabis and increased potency

In recent years, the use of medical cannabis, which is used to address adverse effects of chemotherapy as well as neuropathic pain, Parkinson’s disease, and epilepsy, has been increasing.13 However, there is a lack of well-conducted randomized clinical trials evaluating medical cannabis’ efficacy and safety. As medical cannabis continues to gain public acceptance and more states permit its legal use, patients and physicians should be fully informed of the known adverse effects, including impaired attention, learning, and motivation.13

Several studies have drawn attention to the dose-dependence of many of cannabis’ effects. Since at least the 1960s, the concentration of THC in cannabis has increased substantially, thus increasing its potency. Based on 66,747 samples across 8 studies, 1 meta-analysis estimated that THC concentrations in herbal cannabis increased by 0.29% (P < .001) each year between 1970 and 2017.14 Similarly, THC concentrations in cannabis resins were found to have increased by 0.57% (P = .017) each year between 1975 and 2017.14 Cannabis products with high concentrations of THC carry an increased risk of addiction and mental health disorders.14

 

Identifying those at highest risk

Despite ongoing research, scientific consensus on the relationship of cannabis to schizophrenia and psychosis has yet to be reached. The disparity between the relatively high prevalence of regular adult use of cannabis (8%7)and the low incidence of cannabis-induced psychosis suggests that cannabis use alone is unlikely to lead to episodes of psychosis in individuals who are not predisposed to such episodes. Sarrazin et al15 evaluated 170 patients with schizophrenia, 31 of whom had cannabis use disorder. They found no significant difference in lifetime symptom dimensions between groups, and proposed that cannabis-associated schizophrenia should not be categorized as a distinct clinical entity of schizophrenia with specific features.15

Policies that encourage follow-up of patients after episodes of drug-induced psychosis may mitigate the adverse social and economic effects of schizophrenia. Currently, these policies are not widely implemented in health care institutions, possibly because psychotic symptoms may fade after the drug’s effects have dissipated. Despite this, these patients are at high risk of developing schizophrenia and self-harm. New-onset schizophrenia should be promptly identified because delayed diagnosis is associated with worse prognosis.6 Additionally, identifying genetic susceptibilities to schizophrenia—such as the Val158Met polymorphisms—in individuals who use cannabis could help clinicians manage or slow the onset or progression of schizophrenia.3 Motivational interviewing strategies should be used to minimize or eliminate cannabis use in individuals with active schizophrenia or psychosis, thus preventing worse outcomes.

Bottom Line

Identifying susceptibilities to schizophrenia may guide interventions in patients who use cannabis. Several large studies have suggested that cannabis use may exacerbate symptoms and worsen the prognosis of schizophrenia. Motivational interviewing strategies aimed at minimizing cannabis use may improve outcomes in patients with schizophrenia.

Related Resources

  • Khokhar JY, Dwiel LL, Henricks AM, et al. The link between schizophrenia and substance use disorder: a unifying hypothesis. Schizophr Res. 2018;194:78-85. doi:10.1016/j. schres.2017.04.016
  • Otite ES, Solanky A, Doumas S. Adolescents, THC, and the risk of psychosis. Current Psychiatry. 2021;20(12):e1-e2. doi:10.12788/cp.0197

[embed:render:related:node:249151]

References

1. Simeone JC, Ward AJ, Rotella P, et al. An evaluation of variation in published estimates of schizophrenia prevalence from 1990-2013: a systematic literature review. BMC Psychiatry. 2015;15(1):193. doi:10.1186/s12888-015-0578-7

2. Tandon R, Gaebel W, Barch DM, et al. Definition and description of schizophrenia in the DSM-5. Schizophr Res. 2013;150(1):3-10. doi:10.1016/j.schres.2013.05.028

3. Bosia M, Buonocore M, Bechi M, et al. Schizophrenia, cannabis use and catechol-O-methyltransferase (COMT): modeling the interplay on cognition. Prog Neuropsychopharmacol Biol Psychiatry. 2019;92:363-368. doi:10.1016/j.pnpbp.2019.02.009

4. Welch KA, McIntosh AM, Job DE, et al. The impact of substance use on brain structure in people at high risk of developing schizophrenia. Schizophr Bull. 2011;37(5):1066-1076. doi:10.1093/schbul/sbq013

5. Winship IR, Dursun SM, Baker GB, et al. An overview of animal models related to schizophrenia. Can J Psychiatry. 2019;64(1):5-17. doi:10.1177/0706743718773728

6. Starzer MSK, Nordentoft M, Hjorthøj C. Rates and predictors of conversion to schizophrenia or bipolar disorder following substance-induced psychosis. Am J Psychiatry. 2018;175(4):343-350. doi:10.1176/appi.ajp.2017.17020223

7. Hall W. Cannabis use and psychosis. Drug Alcohol Rev. 1998;17(4):433-444. doi:10.1080/09595239800187271

8. Misiak B, Frydecka D, Slezak R, et al. Elevated homocysteine level in first-episode schizophrenia patients—the relevance of family history of schizophrenia and lifetime diagnosis of cannabis abuse. Metab Brain Dis. 2014;29(3):661-670. doi:10.1007/s11011-014-9534-3

9. Veen ND, Selten J, van der Tweel I, et al. Cannabis use and age at onset of schizophrenia. Am J Psychiatry. 2004;161(3):501-506. doi:10.1176/appi.ajp.161.3.501

10. Foti DJ, Kotov R, Guey LT, et al. Cannabis use and the course of schizophrenia: 10-year follow-up after first hospitalization. Am J Psychiatry. 2010;167(8):987-993. doi:10.1176/appi.ajp.2010.09020189

11. Ortiz-Medina MB, Perea M, Torales J, et al. Cannabis consumption and psychosis or schizophrenia development. Int J Soc Psychiatry. 2018;64(7):690-704. doi:10.1177/0020764018801690

12. Hickman M, Vickerman P, Macleod J, et al. If cannabis caused schizophrenia—how many cannabis users may need to be prevented in order to prevent one case of schizophrenia? England and Wales calculations. Addiction. 2009;104(11):1856-1861. doi:10.1111/j.1360-0443.2009.02736.x

13. Gupta S, Phalen T, Gupta S. Medical marijuana: do the benefits outweigh the risks? Current Psychiatry. 2018;17(1):34-41.

14. Freeman TP, Craft S, Wilson J, et al. Changes in delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) concentrations in cannabis over time: systematic review and meta-analysis. Addiction. 2021;116(5):1000-1010. doi:10.1111/add.15253

15. Sarrazin S, Louppe F, Doukhan R, et al. A clinical comparison of schizophrenia with and without pre-onset cannabis use disorder: a retrospective cohort study using categorical and dimensional approaches. Ann Gen Psychiatry. 2015;14:44. doi:10.1186/s12991-015-0083-x

References

1. Simeone JC, Ward AJ, Rotella P, et al. An evaluation of variation in published estimates of schizophrenia prevalence from 1990-2013: a systematic literature review. BMC Psychiatry. 2015;15(1):193. doi:10.1186/s12888-015-0578-7

2. Tandon R, Gaebel W, Barch DM, et al. Definition and description of schizophrenia in the DSM-5. Schizophr Res. 2013;150(1):3-10. doi:10.1016/j.schres.2013.05.028

3. Bosia M, Buonocore M, Bechi M, et al. Schizophrenia, cannabis use and catechol-O-methyltransferase (COMT): modeling the interplay on cognition. Prog Neuropsychopharmacol Biol Psychiatry. 2019;92:363-368. doi:10.1016/j.pnpbp.2019.02.009

4. Welch KA, McIntosh AM, Job DE, et al. The impact of substance use on brain structure in people at high risk of developing schizophrenia. Schizophr Bull. 2011;37(5):1066-1076. doi:10.1093/schbul/sbq013

5. Winship IR, Dursun SM, Baker GB, et al. An overview of animal models related to schizophrenia. Can J Psychiatry. 2019;64(1):5-17. doi:10.1177/0706743718773728

6. Starzer MSK, Nordentoft M, Hjorthøj C. Rates and predictors of conversion to schizophrenia or bipolar disorder following substance-induced psychosis. Am J Psychiatry. 2018;175(4):343-350. doi:10.1176/appi.ajp.2017.17020223

7. Hall W. Cannabis use and psychosis. Drug Alcohol Rev. 1998;17(4):433-444. doi:10.1080/09595239800187271

8. Misiak B, Frydecka D, Slezak R, et al. Elevated homocysteine level in first-episode schizophrenia patients—the relevance of family history of schizophrenia and lifetime diagnosis of cannabis abuse. Metab Brain Dis. 2014;29(3):661-670. doi:10.1007/s11011-014-9534-3

9. Veen ND, Selten J, van der Tweel I, et al. Cannabis use and age at onset of schizophrenia. Am J Psychiatry. 2004;161(3):501-506. doi:10.1176/appi.ajp.161.3.501

10. Foti DJ, Kotov R, Guey LT, et al. Cannabis use and the course of schizophrenia: 10-year follow-up after first hospitalization. Am J Psychiatry. 2010;167(8):987-993. doi:10.1176/appi.ajp.2010.09020189

11. Ortiz-Medina MB, Perea M, Torales J, et al. Cannabis consumption and psychosis or schizophrenia development. Int J Soc Psychiatry. 2018;64(7):690-704. doi:10.1177/0020764018801690

12. Hickman M, Vickerman P, Macleod J, et al. If cannabis caused schizophrenia—how many cannabis users may need to be prevented in order to prevent one case of schizophrenia? England and Wales calculations. Addiction. 2009;104(11):1856-1861. doi:10.1111/j.1360-0443.2009.02736.x

13. Gupta S, Phalen T, Gupta S. Medical marijuana: do the benefits outweigh the risks? Current Psychiatry. 2018;17(1):34-41.

14. Freeman TP, Craft S, Wilson J, et al. Changes in delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) concentrations in cannabis over time: systematic review and meta-analysis. Addiction. 2021;116(5):1000-1010. doi:10.1111/add.15253

15. Sarrazin S, Louppe F, Doukhan R, et al. A clinical comparison of schizophrenia with and without pre-onset cannabis use disorder: a retrospective cohort study using categorical and dimensional approaches. Ann Gen Psychiatry. 2015;14:44. doi:10.1186/s12991-015-0083-x

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
43-47
Page Number
43-47
Publications
Publications
Topics
Article Type
Display Headline
Cannabis and schizophrenia: A complex relationship
Display Headline
Cannabis and schizophrenia: A complex relationship
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1223 Maes</fileName> <TBEID>0C02E986.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02E986</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Cannabis and schizophrenia:&#13;A c</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231129T080954</firstPublished> <LastPublished>20231129T080954</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231129T080954</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Joseph M. Maes, BS, Charlotte A. Ritchie, MD</byline> <bylineText/> <bylineFull>Joseph M. Maes, BS, Charlotte A. Ritchie, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>43-47</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Approximately 1 in 200 individuals will be diagnosed with schizophrenia in their lifetime.1 DSM-5 criteria for the diagnosis of schizophrenia require the presen</metaDescription> <articlePDF>299299</articlePDF> <teaserImage/> <title>Cannabis and schizophrenia: A complex relationship</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords> <keyword>schizophrenia</keyword> <keyword> addiction medicine</keyword> <keyword> cannabis</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">26935</term> </sections> <topics> <term canonical="true">293</term> <term>174</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002646.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Cannabis and schizophrenia: A complex relationship</title> <deck/> </itemMeta> <itemContent> <p class="abstract">For some who are predisposed to psychosis, cannabis may trigger or exacerbate symptoms</p> <p>Approximately 1 in 200 individuals will be diagnosed with schizophrenia in their lifetime.<sup>1</sup> DSM-5 criteria for the diagnosis of schizophrenia require the presence of ≥2 of 5 symptoms: delusions, hallucinations, disordered speech, grossly disorganized (or catatonic) behavior, and negative symptoms such as flat affect or avolition.<sup>2</sup> Multiple studies have found increased rates of cannabis use among patients with schizophrenia. Because cognitive deficits are the chief predictor of clinical outcomes and quality of life in individuals with schizophrenia, the cognitive effects of cannabis use among these patients are of clinical significance.<sup>3</sup> As legislation increasingly allows for the sale, possession, and consumption of cannabis, it is crucial to provide clinicians with evidence-based recommendations for treating patients who regularly use cannabis (approximately 8% of the adult population<sup>3</sup>). In this article, we analyze several peer-reviewed studies to investigate the impact of cannabis use on the onset and development of schizophrenia.</p> <h3>A look at substance-induced psychosis</h3> <p>Schizophrenia is associated with several structural brain changes, and some of these changes may be influenced by cannabis use (<span class="bitalic">Box</span><i>,</i><sup>4 </sup><span class="bitalic">page 44</span>). The biochemical etiology of schizophrenia is poorly understood but thought to involve dopamine, glutamate, serotonin, and gamma-aminobutyric acid. Certain positive symptoms, such as hallucinations, are uniquely human and difficult to study in animal models.<sup>5</sup> Psychoactive substance use, especially cannabis, is frequently comorbid with schizophrenia. Additionally, certain individuals may be more predisposed to substance-induced psychosis than others based on genetic variation and underlying brain structure changes.<sup>4</sup> Substance-induced psychosis is a psychotic state following the ingestion of a psychoactive substance or drug withdrawal lasting ≥48 hours.<sup>6</sup> The psychoactive effects of cannabis have been associated with an exacerbation of existing schizophrenia symptoms.<sup>7</sup> In 1998, Hall<sup>7</sup> proposed 2 hypotheses to explain the relationship between cannabis and psychosis. The first was that heavy consumption of cannabis triggers a specific type of cannabis psychosis.<sup>7</sup> The second was that cannabis use exacerbates existing schizophrenia, making the symptoms worse.<sup>7</sup> Hall<sup>7</sup> concluded that there was a complicated interaction among an individual’s vulnerability to their stressors, environment, and genetics.</p> <h3>Cannabis, COMT, and homocysteine</h3> <p>Great advances have been made in our ability to examine the association between genetics and metabolism. One example of this is the interaction between the catechol-O-methyltransferase (COMT) gene and the active component of cannabis, delta-9-tetra­hydrocannabinol (THC). COMT codes for an enzyme that degrades cortical dopamine. The Val158Met polymorphism of this gene increases COMT activity, leading to increased dopamine catabolism, and thus decreased levels of extracellular dopamine, which induces an increase in mesolimbic dopaminergic activity, thereby increasing susceptibility to psychosis.<sup>3</sup></p> <p>In a study that genotyped 135 patients with schizophrenia, the Val158Met polymorphism was present in 29.63% of participants.<sup>3</sup> Because THC can induce episodes of psychosis, individuals with this polymorphism may be at a higher risk of developing schizophrenia. Compared to Met carrier control participants with similar histories of cannabis consumption, those with the Val158Met polymorphism demonstrated markedly worse performance on tests of verbal fluency and processing speed.<sup>3</sup> This is clinically significant because cognitive impairments are a major prognostic factor in schizophrenia, and identifying patients with this polymorphism could help personalize interventions for those who consume cannabis and are at risk of developing schizophrenia.<br/><br/>A study that evaluated 56 patients with first-episode schizophrenia found that having a history of cannabis abuse was associated with significantly higher levels of homocysteine as well as lower levels of high-density lipoprotein and vitamin B12.<sup>8</sup> Homocysteine is an agonist at the glutamate binding site and a partial antagonist at the glycine co-agonist site in the <i>N</i>-methyl-<scaps>d</scaps>-aspartate receptor, which suggests that homocysteine may contribute to hypofunctioning of glutamate transmission; this is implicated in the development of schizophrenia. These increases in homocysteine are also found in siblings of patients with schizophrenia, which indicates a possible association between the methylenetetrahydrofolate (MTHFR) gene and schizophrenia.<sup>8</sup>The C677T polymorphism in MTHFR may predict the risk of developing metabolic syndrome in patients taking second-generation antipsychotics.<sup>8</sup> Elevations in homocysteine by as little as 5 μmol/L may increase schizophrenia risk by 70% compared to controls, possibly due to homocysteine initiating neuronal apoptosis, catalyzing dysfunction of the mitochondria, or increasing oxidative stress.<sup>8</sup> There is a positive correlation between homocysteine levels and severity of negative symptoms (<i>P</i> = .006) and general psychopathology (<i>P</i> = .008) of schizophrenia when analyzed using the Positive and Negative Syndrome Scale.<sup>8</sup> Negative symptoms such as blunted affect, apathy, anhedonia, and loss of motivation significantly impact the social and economic outcomes of patients diagnosed with schizophrenia.</p> <h3>Research paints a mixed picture</h3> <p>A Danish study analyzed the rates of conversion to schizophrenia or bipolar disorder (BD) among 6,788 individuals who received a diagnosis of substance-induced psychosis from 1994 to 2014.<sup>6</sup> Ten comparison participants were selected for each case participant, matched on sex and year/month of birth. Participants were followed until the first occurrence of schizophrenia or BD, death, or emigration from Denmark. Substances implicated in the initial psychotic episode included cannabis, alcohol, opioids, sedatives, cocaine, amphetamines, hallucinogens, and combinations of substances.</p> <p>The overall conversion rate over 20 years was 32.2% (95% CI, 29.7 to 34.9), with 26.0% developing schizophrenia vs 8.4% developing BD.<sup>6</sup> Of the substances involved, cannabis was the most common, implicated in 41.2% (95% CI, 36.6 to 46.2) of cases.<sup>6</sup> One-half of male patients converted within 2.0 years and one-half of female patients converted within 4.4 years after a cannabis-induced psychosis.<sup>6</sup><br/><br/>This study had several limitations. It could not account for any short-term psychotic symptoms experienced by the general population, especially after cannabis use. Such patients might not seek treatment. Thus, the results might not be generalizable to the general population. The study did not evaluate if conversion rates differed based on continued substance use following the psychosis episode, or the amount of each substance taken prior to the episode. Dose-dependence was not well elucidated, and this study only looked at patients from Denmark and did not account for socioeconomic status.<sup>6</sup><br/><br/>Another Danish study looked at the influences of gender and cannabis use in the early course of the disease in 133 patients with schizophrenia.<sup>9</sup> These researchers found that male gender was a significant predictor of earlier onset of dysfunction socially and in the workplace, as well as a higher risk of developing negative symptoms. However, compared to gender, cannabis use was a stronger predictor of age at first psychotic episode. For cannabis users, the median age of onset of negative symptoms was 23.7, compared to 38.4 for nonusers (<i>P</i> &lt; .001).<sup>9</sup><br/><br/>Cannabis use is significantly elevated among individuals with psychosis, with a 12-month prevalence of 29.2% compared to 4.0% among the general population of the United States.<sup>10</sup> In a study that assessed 229 patients with a schizophrenia spectrum disorder during their first hospitalization and 6 months, 2 years, 4 years, and 10 years later, Foti et al<sup>10</sup> found that the lifetime rate of cannabis use was 66.2%. Survival analysis found cannabis use doubled the risk of the onset of psychosis compared to nonusers of the same age (hazard ratio [HR] = 1.97; 95% CI, 1.48 to 2.62, <i>P</i> &lt; .001), even after adjusting for socioeconomic status, age, and gender (HR = 1.34; 95% CI, 1.01 to 1.77, <i>P</i> &lt; .05).<sup>10</sup> Additionally, Foti et al<sup>10</sup> found significant positive correlations between psychotic symptoms and cannabis use in patients with schizophrenia over the course of 10 years. An increase in symptoms was associated with a higher likelihood of cannabis use, and a decrease in symptoms was correlated with a lower likelihood of use (adjusted odds ratio = 1.64; 95% CI, 1.12 to 2.43, <i>P</i> &lt; .0125).<sup>10</sup><br/><br/>Ortiz-Medina et al<sup>11</sup> conducted a meta-analysis of 22 studies of 15 cohorts from healthy populations and 12 other cohort follow-up studies that evaluated the onset of psychotic symptoms in individuals who used cannabis. Most studies found associations between cannabis use and the onset of symptoms of schizophrenia, and most determined cannabis was also a major risk factor for other psychotic disorders. Analyses of dose-dependence indicated that repeated cannabis use increased the risk of developing psychotic symptoms. This risk is increased when an individual starts using cannabis before age 15.<sup>11</sup> Age seemed to be a stronger predictor of onset and outcome than sex, with no significant differences between men and women. One study in this review found that approximately 8% to 13% cases of schizophrenia may have been solely due to cannabis.<sup>11</sup> The most significant limitation to the studies analyzed in this review is that retrospective studies utilize self-reported questionnaires.<br/><br/>Other researchers have found it would take a relatively high number of individuals to stop using cannabis to prevent 1 case of schizophrenia. In a study of data from England and Wales, Hickman et al<sup>12</sup> evaluated the best available estimates of the incidence of schizophrenia, rates of heavy and light cannabis use, and risk that cannabis causes schizophrenia to determine the number needed to prevent (NNP) 1 case of schizophrenia. They estimated that it would require approximately 2,800 men age 20 to 24 (90% CI, 2,018 to 4,530) and 4,700 men age 35 to 39 (90% CI, 3,114 to 8,416) who heavily used cannabis to stop their consumption to prevent 1 case of schizophrenia.<sup>12</sup> For women with heavy cannabis use, the mean NNP was 5,470 for women age 25 to 29 (90% CI, 3,640 to 9,839) and 10,870 for women age 35 to 39 (90% CI, 6,786 to 22,732).<sup>12</sup> For light cannabis users, the NNP was 4 to 5 times higher than the NNP for heavy cannabis users. This suggests that clinical interventions aimed at preventing dependence on cannabis would be more effective than interventions aimed at eliminating cannabis use.</p> <h3>Medical cannabis and increased potency</h3> <p>In recent years, the use of medical cannabis, which is used to address adverse effects of chemotherapy as well as neuropathic pain, Parkinson’s disease, and epilepsy, has been increasing.<sup>13</sup> However, there is a lack of well-conducted randomized clinical trials evaluating medical cannabis’ efficacy and safety. As medical cannabis continues to gain public acceptance and more states permit its legal use, patients and physicians should be fully informed of the known adverse effects, including impaired attention, learning, and motivation.<sup>13</sup></p> <p>Several studies have drawn attention to the dose-dependence of many of cannabis’ effects. Since at least the 1960s, the concentration of THC in cannabis has increased substantially, thus increasing its potency. Based on 66,747 samples across 8 studies, 1 meta-analysis estimated that THC concentrations in herbal cannabis increased by 0.29% (<i>P</i> &lt; .001) each year between 1970 and 2017.<sup>14</sup> Similarly, THC concentrations in cannabis resins were found to have increased by 0.57% (<i>P =</i> .017) each year between 1975 and 2017.<sup>14</sup> Cannabis products with high concentrations of THC carry an increased risk of addiction and mental health disorders.<sup>14</sup></p> <h3>Identifying those at highest risk</h3> <p>Despite ongoing research, scientific consensus on the relationship of cannabis to schizophrenia and psychosis has yet to be reached. The disparity between the relatively high prevalence of regular adult use of cannabis (8%<sup>7</sup>)<sup> </sup>and the low incidence of cannabis-induced psychosis suggests that cannabis use alone is unlikely to lead to episodes of psychosis in individuals who are not predisposed to such episodes. Sarrazin et al<sup>15</sup> evaluated 170 patients with schizophrenia, 31 of whom had cannabis use disorder. They found no significant difference in lifetime symptom dimensions between groups, and proposed that cannabis-associated schizophrenia should not be categorized as a distinct clinical entity of schizophrenia with specific features.<sup>15</sup></p> <p>Policies that encourage follow-up of patients after episodes of drug-induced psychosis may mitigate the adverse social and economic effects of schizophrenia. Currently, these policies are not widely implemented in health care institutions, possibly because psychotic symptoms may fade after the drug’s effects have dissipated. Despite this, these patients are at high risk of developing schizophrenia and self-harm. New-onset schizophrenia should be promptly identified because delayed diagnosis is associated with worse prognosis.<sup>6</sup> Additionally, identifying genetic susceptibilities to schizophrenia—such as the Val158Met polymorphisms—in individuals who use cannabis could help clinicians manage or slow the onset or progression of schizophrenia.<sup>3</sup> Motivational interviewing strategies should be used to minimize or eliminate cannabis use in individuals with active schizophrenia or psychosis, thus preventing worse outcomes.</p> <p> <strong>References</strong> </p> <p class="reference"> 1. Simeone JC, Ward AJ, Rotella P, et al. An evaluation of variation in published estimates of schizophrenia prevalence from 1990-2013: a systematic literature review. BMC Psychiatry. 2015;15(1):193. doi:10.1186/s12888-015-0578-7<br/><br/> 2. Tandon R, Gaebel W, Barch DM, et al. Definition and description of schizophrenia in the DSM-5. Schizophr Res. 2013;150(1):3-10. doi:10.1016/j.schres.2013.05.028<br/><br/> 3. Bosia M, Buonocore M, Bechi M, et al. Schizophrenia, cannabis use and catechol-O-methyltransferase (COMT): modeling the interplay on cognition. Prog Neuropsychopharmacol Biol Psychiatry. 2019;92:363-368. doi:10.1016/j.pnpbp.2019.02.009<br/><br/> 4. Welch KA, McIntosh AM, Job DE, et al. The impact of substance use on brain structure in people at high risk of developing schizophrenia. Schizophr Bull. 2011;37(5):1066-1076. doi:10.1093/schbul/sbq013<br/><br/> 5. Winship IR, Dursun SM, Baker GB, et al. An overview of animal models related to schizophrenia. Can J Psychiatry. 2019;64(1):5-17. doi:10.1177/0706743718773728<br/><br/> 6. Starzer MSK, Nordentoft M, Hjorthøj C. Rates and predictors of conversion to schizophrenia or bipolar disorder following substance-induced psychosis. Am J Psychiatry. 2018;175(4):343-350. doi:10.1176/appi.ajp.2017.17020223<br/><br/> 7. Hall W. Cannabis use and psychosis. Drug Alcohol Rev. 1998;17(4):433-444. doi:10.1080/09595239800187271<br/><br/> 8. Misiak B, Frydecka D, Slezak R, et al. Elevated homocysteine level in first-episode schizophrenia patients—the relevance of family history of schizophrenia and lifetime diagnosis of cannabis abuse. Metab Brain Dis. 2014;29(3):661-670. doi:10.1007/s11011-014-9534-3<br/><br/> 9. Veen ND, Selten J, van der Tweel I, et al. Cannabis use and age at onset of schizophrenia. Am J Psychiatry. 2004;161(3):501-506. doi:10.1176/appi.ajp.161.3.501<br/><br/> 10. Foti DJ, Kotov R, Guey LT, et al. Cannabis use and the course of schizophrenia: 10-year follow-up after first hospitalization. Am J Psychiatry. 2010;167(8):987-993. doi:10.1176/appi.ajp.2010.09020189</p> <p class="reference"> 11. Ortiz-Medina MB, Perea M, Torales J, et al. Cannabis consumption and psychosis or schizophrenia development. Int J Soc Psychiatry. 2018;64(7):690-704. doi:10.1177/0020764018801690</p> <p class="reference"> 12. Hickman M, Vickerman P, Macleod J, et al. If cannabis caused schizophrenia—how many cannabis users may need to be prevented in order to prevent one case of schizophrenia? England and Wales calculations. Addiction. 2009;104(11):1856-1861. doi:10.1111/j.1360-0443.2009.02736.x<br/><br/> 13. Gupta S, Phalen T, Gupta S. Medical marijuana: do the benefits outweigh the risks? Current Psychiatry. 2018;17(1):34-41.<br/><br/> 14. Freeman TP, Craft S, Wilson J, et al. Changes in delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) concentrations in cannabis over time: systematic review and meta-analysis. Addiction. 2021;116(5):1000-1010. doi:10.1111/add.15253<br/><br/> 15. Sarrazin S, Louppe F, Doukhan R, et al. A clinical comparison of schizophrenia with and without pre-onset cannabis use disorder: a retrospective cohort study using categorical and dimensional approaches. Ann Gen Psychiatry. 2015;14:44. doi:10.1186/s12991-015-0083-x</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002646.SIG
Disable zoom
Off

Shakespeare and suicide

Article Type
Changed
Wed, 12/06/2023 - 10:06
Display Headline
Shakespeare and suicide

The medical knowledge that William Shakespeare possessed has awed scholars for centuries. Theories about the provenance of his knowledge abound (such as his son-in-law being a physician), and the inclusion of medical terms and ailments throughout his plays suggests a broad knowledge of disease and sickness. Scholars have noted how he sprinkles references to dermatologic, neurologic, orthopedic, and metabolic ailments throughout his plays, mentioning carbuncles, fistulas, corpulence, rhinophyma, scurvy, ague, enuresis, kyphosis, epilepsy, and parkinsonism.1 What seems to strike post-Enlightenment audiences—and what sets Shakespeare apart from many of his contemporaries—is his portrayal of “complex” characters, those with what we envision as rich interior worlds and with whom a modern audience can resonate. There is a reason psychiatrists such as Sigmund Freud have rushed back to Shakespeare and (sometimes anachronistically) found in his characters various psychiatric diagnoses such as depression, anxiety, paranoia, jealous delusions, and obsessive-compulsive disorder. Suicide and suicidal ideation are prevalent themes in some of Shakespeare’s most well-known characters.

A surprisingly common theme

The gravest outcome of a psychiatric illness is death by suicide, which occurs in 13 of Shakespeare’s characters.2 There are additional characters who exhibit suicidal ideation without a completed act. Shakespearean characters whose lives end in suicide are variably portrayed, dying by various means and circumstances. Hamlet (who dies at the hand of his foe, Laertes), famously soliloquizes the theme of suicide and the afterlife. He ponders “tak[ing] arms against a sea of troubles.” Ophelia dies ambiguously. Immediately after, her mother and brother recount her death in a brook—having had “too much of water” when her garments “heavy with their drink, | pull’d the poor wretch from her melodious lay | To muddy death.” The 2 clowns/gravediggers then debate whether Ophelia deserves a Christian burial and if her death should be considered a suicide: did the water drown her, or did she drown herself?3

Lady Macbeth’s suicide is offstage, punctuated by a “night-shriek.” Romeo drinks poison and dies “with a kiss.” Juliet quickly follows, making her body the sword’s sheath which “there rust, and let [her] die.” Othello stabs himself after requesting that his peers will “speak of me as I am.” One of King Lear’s daughters poisons her sister “and after [slays] herself.” Timon dies by his cave, “entomb’d upon the very hem o’ the sea.” In Antony and Cleopatra, after being told that Cleopatra has killed herself with Antony’s name on her lips, Antony begs to be stabbed and then stabs himself; he is not defeated by Caesar, but rather conquered by himself: “none but Antony | Should conquer Antony.” Cleopatra and her lady-in-waiting, Charmian, kill themselves with an asp. In Julius Caesar, Brutus runs upon his sword. Cassius begs for his own death, asking that “this good sword, | That ran through Caesar’s bowels, search this bosom.” Portia, it is reported, “swallowed fire.”

Shakespeare uses specific stylized language to portray characters in psychological anguish and suicidal states. Scholars have discussed how he uses certain stylistic language to highlight the anguish that happens during solitary, solipsistic moments of contemplation.4 Moments of anguish and suicidal ideation are marked by verbal repetition. An example of this repetition comes in Hamlet’s speech after he returns to the kingdom where his uncle has usurped his father, when he laments that he cannot end his own life. He says:

O, that this too too sullied flesh would melt,
Thaw, and resolve itself into a dew!
Or that the Everlasting had not fix’d
His canon ’gainst self-slaughter! O God, God,
How weary, stale, flat, and unprofitable
Seem to me all the uses of this world.

In these 6 lines, there are 2 instances of verbal repetition: “too too” and “God, God.” In this moment of solitude and despair, Hamlet’s speech fractures; his fractured speech reflects his fractured psyche. While Hamlet speaks of staleness and stagnation in the world, his words represent a sterile excess. No meaning is elicited by their repetition; there is no forward momentum to his speech. The words reflect the extent to which Hamlet is stuck and divided in this moment. Something similar happens in Macbeth’s “Tomorrow and tomorrow and tomorrow” speech. The words march on, and with each repetition they become increasingly hollow and brittle.

Why does this discussion of suicide in Shakespeare hold value for a contemporary psychiatrist? First, there is no single prototypical suicidal character in Shakespeare. His characters who are suicidal vary in their demographics and incentives for ending their lives. In this way, he provides a rich framework, one with which many people can engage. Second, this discussion fits into an existing paradigm for using art therapy (specifically Shakespeare) as a treatment modality for trauma.5 Programs such as DE-CRUIT have used the recitation of Shakespearean verse as a means of processing trauma in veterans.5 While Shakespeare does not mention a remedy for suicide in his plays, perhaps the text can serve as medicine. Third, the repetitive speech that Shakespeare uses in times of anguish could be a fairly accurate reflection of speech patterns in patients who are suicidal. Research that completed a spoken language analysis of patients who were suicidal has found “mechanical and repetitive phrasing” as a quality of these patients’ speech.6,7

For hundreds of years, critics have searched beyond the text for Shakespeare’s voice and opinion; what did he himself think of melancholy, despair, or suicide? We cannot know. We, as readers, are invited to explore a nuanced and multifaceted view of suicide, one that neither chides nor valorizes the act, and provides ambiguity rather than condemnation.

References

1. Paciaroni M, Bogousslavsky J. William Shakespeare’s neurology. Prog Brain Res. 2013;206:3-18.

2. Kirkland LR. To end itself by death: suicide in Shakespeare’s tragedies. South Med J. 1999;92(7):660-666.

3. Sale C. The “Amending Hand”: Hales v. Petit, Eyston v. Studd, and Equitable Action in Hamlet. In: Jordan C, Cunningham K, eds. The Law in Shakespeare. Early Modern Literature in History. Palgrave Macmillan; 2007:189-207. https://doi.org/10.1057/9780230626348_11

4. Langley E. Narcissism and Suicide in Shakespeare and His Contemporaries. Oxford University Press; 2009.

5. Ali A, Wolfert S, Lam I, et al. Intersecting modes of aesthetic distance and mimetic induction in therapeutic process: examining a drama-based treatment for military-related traumatic stress. Drama Therapy Review. 2018;4(2):153-165.

6. Belouali A, Gupta S, Sourirajan V, et al. Acoustic and language analysis of speech for suicidal ideation among US veterans. BioData Min. 2021;14(1):11. doi:10.1186/s13040-021-00245-y

7. Cummins N, Scherer S, Krajewski J, et al. A review of depression and suicide risk assessment using speech analysis. Speech Commun. 2015;71:10-49.

Article PDF
Author and Disclosure Information

Susanna Gebhardt, PhD
Medical Student
University of Cincinnati College of Medicine
Cincinnati, Ohio

Henry A. Nasrallah, MD, DLFAPA
Professor of Psychiatry, Neurology, and Neuroscience
Department of Psychiatry and Behavioral Neuroscience
University of Cincinnati College of Medicine
Cincinnati, Ohio

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Issue
Current Psychiatry - 22(12)
Publications
Page Number
22-24
Sections
Author and Disclosure Information

Susanna Gebhardt, PhD
Medical Student
University of Cincinnati College of Medicine
Cincinnati, Ohio

Henry A. Nasrallah, MD, DLFAPA
Professor of Psychiatry, Neurology, and Neuroscience
Department of Psychiatry and Behavioral Neuroscience
University of Cincinnati College of Medicine
Cincinnati, Ohio

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Author and Disclosure Information

Susanna Gebhardt, PhD
Medical Student
University of Cincinnati College of Medicine
Cincinnati, Ohio

Henry A. Nasrallah, MD, DLFAPA
Professor of Psychiatry, Neurology, and Neuroscience
Department of Psychiatry and Behavioral Neuroscience
University of Cincinnati College of Medicine
Cincinnati, Ohio

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Article PDF
Article PDF

The medical knowledge that William Shakespeare possessed has awed scholars for centuries. Theories about the provenance of his knowledge abound (such as his son-in-law being a physician), and the inclusion of medical terms and ailments throughout his plays suggests a broad knowledge of disease and sickness. Scholars have noted how he sprinkles references to dermatologic, neurologic, orthopedic, and metabolic ailments throughout his plays, mentioning carbuncles, fistulas, corpulence, rhinophyma, scurvy, ague, enuresis, kyphosis, epilepsy, and parkinsonism.1 What seems to strike post-Enlightenment audiences—and what sets Shakespeare apart from many of his contemporaries—is his portrayal of “complex” characters, those with what we envision as rich interior worlds and with whom a modern audience can resonate. There is a reason psychiatrists such as Sigmund Freud have rushed back to Shakespeare and (sometimes anachronistically) found in his characters various psychiatric diagnoses such as depression, anxiety, paranoia, jealous delusions, and obsessive-compulsive disorder. Suicide and suicidal ideation are prevalent themes in some of Shakespeare’s most well-known characters.

A surprisingly common theme

The gravest outcome of a psychiatric illness is death by suicide, which occurs in 13 of Shakespeare’s characters.2 There are additional characters who exhibit suicidal ideation without a completed act. Shakespearean characters whose lives end in suicide are variably portrayed, dying by various means and circumstances. Hamlet (who dies at the hand of his foe, Laertes), famously soliloquizes the theme of suicide and the afterlife. He ponders “tak[ing] arms against a sea of troubles.” Ophelia dies ambiguously. Immediately after, her mother and brother recount her death in a brook—having had “too much of water” when her garments “heavy with their drink, | pull’d the poor wretch from her melodious lay | To muddy death.” The 2 clowns/gravediggers then debate whether Ophelia deserves a Christian burial and if her death should be considered a suicide: did the water drown her, or did she drown herself?3

Lady Macbeth’s suicide is offstage, punctuated by a “night-shriek.” Romeo drinks poison and dies “with a kiss.” Juliet quickly follows, making her body the sword’s sheath which “there rust, and let [her] die.” Othello stabs himself after requesting that his peers will “speak of me as I am.” One of King Lear’s daughters poisons her sister “and after [slays] herself.” Timon dies by his cave, “entomb’d upon the very hem o’ the sea.” In Antony and Cleopatra, after being told that Cleopatra has killed herself with Antony’s name on her lips, Antony begs to be stabbed and then stabs himself; he is not defeated by Caesar, but rather conquered by himself: “none but Antony | Should conquer Antony.” Cleopatra and her lady-in-waiting, Charmian, kill themselves with an asp. In Julius Caesar, Brutus runs upon his sword. Cassius begs for his own death, asking that “this good sword, | That ran through Caesar’s bowels, search this bosom.” Portia, it is reported, “swallowed fire.”

Shakespeare uses specific stylized language to portray characters in psychological anguish and suicidal states. Scholars have discussed how he uses certain stylistic language to highlight the anguish that happens during solitary, solipsistic moments of contemplation.4 Moments of anguish and suicidal ideation are marked by verbal repetition. An example of this repetition comes in Hamlet’s speech after he returns to the kingdom where his uncle has usurped his father, when he laments that he cannot end his own life. He says:

O, that this too too sullied flesh would melt,
Thaw, and resolve itself into a dew!
Or that the Everlasting had not fix’d
His canon ’gainst self-slaughter! O God, God,
How weary, stale, flat, and unprofitable
Seem to me all the uses of this world.

In these 6 lines, there are 2 instances of verbal repetition: “too too” and “God, God.” In this moment of solitude and despair, Hamlet’s speech fractures; his fractured speech reflects his fractured psyche. While Hamlet speaks of staleness and stagnation in the world, his words represent a sterile excess. No meaning is elicited by their repetition; there is no forward momentum to his speech. The words reflect the extent to which Hamlet is stuck and divided in this moment. Something similar happens in Macbeth’s “Tomorrow and tomorrow and tomorrow” speech. The words march on, and with each repetition they become increasingly hollow and brittle.

Why does this discussion of suicide in Shakespeare hold value for a contemporary psychiatrist? First, there is no single prototypical suicidal character in Shakespeare. His characters who are suicidal vary in their demographics and incentives for ending their lives. In this way, he provides a rich framework, one with which many people can engage. Second, this discussion fits into an existing paradigm for using art therapy (specifically Shakespeare) as a treatment modality for trauma.5 Programs such as DE-CRUIT have used the recitation of Shakespearean verse as a means of processing trauma in veterans.5 While Shakespeare does not mention a remedy for suicide in his plays, perhaps the text can serve as medicine. Third, the repetitive speech that Shakespeare uses in times of anguish could be a fairly accurate reflection of speech patterns in patients who are suicidal. Research that completed a spoken language analysis of patients who were suicidal has found “mechanical and repetitive phrasing” as a quality of these patients’ speech.6,7

For hundreds of years, critics have searched beyond the text for Shakespeare’s voice and opinion; what did he himself think of melancholy, despair, or suicide? We cannot know. We, as readers, are invited to explore a nuanced and multifaceted view of suicide, one that neither chides nor valorizes the act, and provides ambiguity rather than condemnation.

The medical knowledge that William Shakespeare possessed has awed scholars for centuries. Theories about the provenance of his knowledge abound (such as his son-in-law being a physician), and the inclusion of medical terms and ailments throughout his plays suggests a broad knowledge of disease and sickness. Scholars have noted how he sprinkles references to dermatologic, neurologic, orthopedic, and metabolic ailments throughout his plays, mentioning carbuncles, fistulas, corpulence, rhinophyma, scurvy, ague, enuresis, kyphosis, epilepsy, and parkinsonism.1 What seems to strike post-Enlightenment audiences—and what sets Shakespeare apart from many of his contemporaries—is his portrayal of “complex” characters, those with what we envision as rich interior worlds and with whom a modern audience can resonate. There is a reason psychiatrists such as Sigmund Freud have rushed back to Shakespeare and (sometimes anachronistically) found in his characters various psychiatric diagnoses such as depression, anxiety, paranoia, jealous delusions, and obsessive-compulsive disorder. Suicide and suicidal ideation are prevalent themes in some of Shakespeare’s most well-known characters.

A surprisingly common theme

The gravest outcome of a psychiatric illness is death by suicide, which occurs in 13 of Shakespeare’s characters.2 There are additional characters who exhibit suicidal ideation without a completed act. Shakespearean characters whose lives end in suicide are variably portrayed, dying by various means and circumstances. Hamlet (who dies at the hand of his foe, Laertes), famously soliloquizes the theme of suicide and the afterlife. He ponders “tak[ing] arms against a sea of troubles.” Ophelia dies ambiguously. Immediately after, her mother and brother recount her death in a brook—having had “too much of water” when her garments “heavy with their drink, | pull’d the poor wretch from her melodious lay | To muddy death.” The 2 clowns/gravediggers then debate whether Ophelia deserves a Christian burial and if her death should be considered a suicide: did the water drown her, or did she drown herself?3

Lady Macbeth’s suicide is offstage, punctuated by a “night-shriek.” Romeo drinks poison and dies “with a kiss.” Juliet quickly follows, making her body the sword’s sheath which “there rust, and let [her] die.” Othello stabs himself after requesting that his peers will “speak of me as I am.” One of King Lear’s daughters poisons her sister “and after [slays] herself.” Timon dies by his cave, “entomb’d upon the very hem o’ the sea.” In Antony and Cleopatra, after being told that Cleopatra has killed herself with Antony’s name on her lips, Antony begs to be stabbed and then stabs himself; he is not defeated by Caesar, but rather conquered by himself: “none but Antony | Should conquer Antony.” Cleopatra and her lady-in-waiting, Charmian, kill themselves with an asp. In Julius Caesar, Brutus runs upon his sword. Cassius begs for his own death, asking that “this good sword, | That ran through Caesar’s bowels, search this bosom.” Portia, it is reported, “swallowed fire.”

Shakespeare uses specific stylized language to portray characters in psychological anguish and suicidal states. Scholars have discussed how he uses certain stylistic language to highlight the anguish that happens during solitary, solipsistic moments of contemplation.4 Moments of anguish and suicidal ideation are marked by verbal repetition. An example of this repetition comes in Hamlet’s speech after he returns to the kingdom where his uncle has usurped his father, when he laments that he cannot end his own life. He says:

O, that this too too sullied flesh would melt,
Thaw, and resolve itself into a dew!
Or that the Everlasting had not fix’d
His canon ’gainst self-slaughter! O God, God,
How weary, stale, flat, and unprofitable
Seem to me all the uses of this world.

In these 6 lines, there are 2 instances of verbal repetition: “too too” and “God, God.” In this moment of solitude and despair, Hamlet’s speech fractures; his fractured speech reflects his fractured psyche. While Hamlet speaks of staleness and stagnation in the world, his words represent a sterile excess. No meaning is elicited by their repetition; there is no forward momentum to his speech. The words reflect the extent to which Hamlet is stuck and divided in this moment. Something similar happens in Macbeth’s “Tomorrow and tomorrow and tomorrow” speech. The words march on, and with each repetition they become increasingly hollow and brittle.

Why does this discussion of suicide in Shakespeare hold value for a contemporary psychiatrist? First, there is no single prototypical suicidal character in Shakespeare. His characters who are suicidal vary in their demographics and incentives for ending their lives. In this way, he provides a rich framework, one with which many people can engage. Second, this discussion fits into an existing paradigm for using art therapy (specifically Shakespeare) as a treatment modality for trauma.5 Programs such as DE-CRUIT have used the recitation of Shakespearean verse as a means of processing trauma in veterans.5 While Shakespeare does not mention a remedy for suicide in his plays, perhaps the text can serve as medicine. Third, the repetitive speech that Shakespeare uses in times of anguish could be a fairly accurate reflection of speech patterns in patients who are suicidal. Research that completed a spoken language analysis of patients who were suicidal has found “mechanical and repetitive phrasing” as a quality of these patients’ speech.6,7

For hundreds of years, critics have searched beyond the text for Shakespeare’s voice and opinion; what did he himself think of melancholy, despair, or suicide? We cannot know. We, as readers, are invited to explore a nuanced and multifaceted view of suicide, one that neither chides nor valorizes the act, and provides ambiguity rather than condemnation.

References

1. Paciaroni M, Bogousslavsky J. William Shakespeare’s neurology. Prog Brain Res. 2013;206:3-18.

2. Kirkland LR. To end itself by death: suicide in Shakespeare’s tragedies. South Med J. 1999;92(7):660-666.

3. Sale C. The “Amending Hand”: Hales v. Petit, Eyston v. Studd, and Equitable Action in Hamlet. In: Jordan C, Cunningham K, eds. The Law in Shakespeare. Early Modern Literature in History. Palgrave Macmillan; 2007:189-207. https://doi.org/10.1057/9780230626348_11

4. Langley E. Narcissism and Suicide in Shakespeare and His Contemporaries. Oxford University Press; 2009.

5. Ali A, Wolfert S, Lam I, et al. Intersecting modes of aesthetic distance and mimetic induction in therapeutic process: examining a drama-based treatment for military-related traumatic stress. Drama Therapy Review. 2018;4(2):153-165.

6. Belouali A, Gupta S, Sourirajan V, et al. Acoustic and language analysis of speech for suicidal ideation among US veterans. BioData Min. 2021;14(1):11. doi:10.1186/s13040-021-00245-y

7. Cummins N, Scherer S, Krajewski J, et al. A review of depression and suicide risk assessment using speech analysis. Speech Commun. 2015;71:10-49.

References

1. Paciaroni M, Bogousslavsky J. William Shakespeare’s neurology. Prog Brain Res. 2013;206:3-18.

2. Kirkland LR. To end itself by death: suicide in Shakespeare’s tragedies. South Med J. 1999;92(7):660-666.

3. Sale C. The “Amending Hand”: Hales v. Petit, Eyston v. Studd, and Equitable Action in Hamlet. In: Jordan C, Cunningham K, eds. The Law in Shakespeare. Early Modern Literature in History. Palgrave Macmillan; 2007:189-207. https://doi.org/10.1057/9780230626348_11

4. Langley E. Narcissism and Suicide in Shakespeare and His Contemporaries. Oxford University Press; 2009.

5. Ali A, Wolfert S, Lam I, et al. Intersecting modes of aesthetic distance and mimetic induction in therapeutic process: examining a drama-based treatment for military-related traumatic stress. Drama Therapy Review. 2018;4(2):153-165.

6. Belouali A, Gupta S, Sourirajan V, et al. Acoustic and language analysis of speech for suicidal ideation among US veterans. BioData Min. 2021;14(1):11. doi:10.1186/s13040-021-00245-y

7. Cummins N, Scherer S, Krajewski J, et al. A review of depression and suicide risk assessment using speech analysis. Speech Commun. 2015;71:10-49.

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
22-24
Page Number
22-24
Publications
Publications
Article Type
Display Headline
Shakespeare and suicide
Display Headline
Shakespeare and suicide
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1223 Gebhardt</fileName> <TBEID>0C02E67E.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02E67E</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Shakespeare and suicide</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231129T081237</firstPublished> <LastPublished>20231129T081237</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231129T081237</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Susanna Gebhardt, PhD, Henry A. Nasrallah, MD, DLFAPA</byline> <bylineText/> <bylineFull>Susanna Gebhardt, PhD, Henry A. Nasrallah, MD, DLFAPA</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>22-24</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>The medical knowledge that William Shakespeare possessed has awed scholars for centuries. Theories about the provenance of his knowledge abound (such as his son</metaDescription> <articlePDF>299291</articlePDF> <teaserImage/> <title>Shakespeare and suicide</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>Decemer</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords> <keyword>suicide</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">52</term> </sections> <topics> <term canonical="true">278</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002642.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Shakespeare and suicide</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Suicide and suicidal ideation are prevalent themes among many Shakespeare characters</p> <p>The medical knowledge that William Shakespeare possessed has awed scholars for centuries. Theories about the provenance of his knowledge abound (such as his son-in-law being a physician), and the inclusion of medical terms and ailments throughout his plays suggests a broad knowledge of disease and sickness. Scholars have noted how he sprinkles references to dermatologic, neurologic, orthopedic, and metabolic ailments throughout his plays, mentioning carbuncles, fistulas, corpulence, rhinophyma, scurvy, ague, enuresis, kyphosis, epilepsy, and parkinsonism.<sup>1</sup> What seems to strike post-Enlightenment audiences—and what sets Shakespeare apart from many of his contemporaries—is his portrayal of “complex” characters, those with what we envision as rich interior worlds and with whom a modern audience can resonate. There is a reason psychiatrists such as Sigmund Freud have rushed back to Shakespeare and (sometimes anachronistically) found in his characters various psychiatric diagnoses such as depression, anxiety, paranoia, jealous delusions, and obsessive-compulsive disorder. Suicide and suicidal ideation are prevalent themes in some of Shakespeare’s most well-known characters.</p> <h3>A surprisingly common theme</h3> <p>The gravest outcome of a psychiatric illness is death by suicide, which occurs in 13 of Shakespeare’s characters.<sup>2</sup> There are additional characters who exhibit suicidal ideation without a completed act. Shakespearean characters whose lives end in suicide are variably portrayed, dying by various means and circumstances. Hamlet (who dies at the hand of his foe, Laertes), famously soliloquizes the theme of suicide and the afterlife. He ponders “tak[ing] arms against a sea of troubles.” Ophelia dies ambiguously. Immediately after, her mother and brother recount her death in a brook—having had “too much of water” when her garments “heavy with their drink, | pull’d the poor wretch from her melodious lay | To muddy death.” The 2 clowns/gravediggers then debate whether Ophelia deserves a Christian burial and if her death should be considered a suicide: did the water drown her, or did she drown herself?<sup>3</sup></p> <p>Lady Macbeth’s suicide is offstage, punctuated by a “night-shriek.” Romeo drinks poison and dies “with a kiss.” Juliet quickly follows, making her body the sword’s sheath which “there rust, and let [her] die.” Othello stabs himself after requesting that his peers will “speak of me as I am.” One of King Lear’s daughters poisons her sister “and after [slays] herself.” Timon dies by his cave, “entomb’d upon the very hem o’ the sea.” In <i>Antony and Cleopatra</i>, after being told that Cleopatra has killed herself with Antony’s name on her lips, Antony begs to be stabbed and then stabs himself; he is not defeated by Caesar, but rather conquered by himself: “none but Antony | Should conquer Antony.” Cleopatra and her lady-in-waiting, Charmian, kill themselves with an asp. In <i>Julius Caesar</i>, Brutus runs upon his sword. Cassius begs for his own death, asking that “this good sword, | That ran through Caesar’s bowels, search this bosom.” Portia, it is reported, “swallowed fire.”<br/><br/>Shakespeare uses specific stylized language to portray characters in psychological anguish and suicidal states. Scholars have discussed how he uses certain stylistic language to highlight the anguish that happens during solitary, solipsistic moments of contemplation.<sup>4</sup> Moments of anguish and suicidal ideation are marked by verbal repetition. An example of this repetition comes in Hamlet’s speech after he returns to the kingdom where his uncle has usurped his father, when he laments that he cannot end his own life. He says:</p> <p> <i>O, that this too too sullied flesh would melt,</i> </p> <p> <i>Thaw, and resolve itself into a dew!<br/><br/>Or that the Everlasting had not fix’d<br/><br/>His canon ’gainst self-slaughter! O God, God,<br/><br/>How weary, stale, flat, and unprofitable</i> </p> <p> <i>Seem to me all the uses of this world.</i> </p> <p>In these 6 lines, there are 2 instances of verbal repetition:<i> </i>“too too” and “God, God.” In this moment of solitude and despair, Hamlet’s speech fractures; his fractured speech reflects his fractured psyche. While Hamlet speaks of staleness and stagnation in the world, his words represent a sterile excess. No meaning is elicited by their repetition; there is no forward momentum to his speech. The words reflect the extent to which Hamlet is stuck and divided in this moment. Something similar happens in Macbeth’s “Tomorrow and tomorrow and tomorrow” speech. The words march on, and with each repetition they become increasingly hollow and brittle.<br/><br/>Why does this discussion of suicide in Shakespeare hold value for a contemporary psychiatrist? First, there is no single prototypical suicidal character in Shakespeare. His characters who are suicidal vary in their demographics and incentives for ending their lives. In this way, he provides a rich framework, one with which many people can engage. Second, this discussion fits into an existing paradigm for using art therapy (specifically Shakespeare) as a treatment modality for trauma.<sup>5</sup> Programs such as DE-CRUIT have used the recitation of Shakespearean verse as a means of processing trauma in veterans.<sup>5</sup> While Shakespeare does not mention a remedy for suicide in his plays, perhaps the text can serve as medicine. Third, the repetitive speech that Shakespeare uses in times of anguish could be a fairly accurate reflection of speech patterns in patients who are suicidal. Research that completed a spoken language analysis of patients who were suicidal has found “mechanical and repetitive phrasing” as a quality of these patients’ speech.<sup>6,7<br/><br/></sup>For hundreds of years, critics have searched beyond the text for Shakespeare’s voice and opinion; what did he himself think of melancholy, despair, or suicide? We cannot know. We, as readers, are invited to explore a nuanced and multifaceted view of suicide, one that neither chides nor valorizes the act, and provides ambiguity rather than condemnation.</p> <p> <strong>References</strong> </p> <p class="reference"> 1. Paciaroni M, Bogousslavsky J. William Shakespeare’s neurology. Prog Brain Res. 2013;206:3-18.<br/><br/> 2. Kirkland LR. To end itself by death: suicide in Shakespeare’s tragedies. South Med J. 1999;92(7):660-666.<br/><br/> 3. Sale C. The “Amending Hand”: Hales v. Petit, Eyston v. Studd, and Equitable Action in Hamlet. In: Jordan C, Cunningham K, eds. <i>The Law in Shakespeare. Early Modern Literature in History.</i> Palgrave Macmillan; 2007:189-207. https://doi.org/10.1057/9780230626348_11<br/><br/> 4. Langley E. <i>Narcissism and Suicide in Shakespeare and His Contemporaries.</i> Oxford University Press; 2009.<br/><br/> 5. Ali A, Wolfert S, Lam I, et al. Intersecting modes of aesthetic distance and mimetic induction in therapeutic process: examining a drama-based treatment for military-related traumatic stress. Drama Therapy Review. 2018;4(2):153-165.<br/><br/> 6. Belouali A, Gupta S, Sourirajan V, et al. Acoustic and language analysis of speech for suicidal ideation among US veterans. BioData Min. 2021;14(1):11. doi:10.1186/s13040-021-00245-y<br/><br/> 7. Cummins N, Scherer S, Krajewski J, et al. A review of depression and suicide risk assessment using speech analysis. Speech Commun. 2015;71:10-49.</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002642.SIG
Disable zoom
Off

Valedictory

Article Type
Changed
Fri, 12/01/2023 - 01:15
Display Headline
Valedictory

All that’s bright must fade,
The brightest still the fleetest;
All that’s sweet was made
But to be lost when sweetest.
Thomas Moore

I sometimes hold it half a sin
To put in words the grief I feel;
For words, like Nature, half reveal
And half conceal the Soul within.
Alfred, Lord Tennyson, In Memoriam

Dear Readers,

I have sad news to share with you. This is the last issue of Current Psychiatry.

During my travels around the country over the past 2 decades, countless psychiatrists have told me that Current Psychiatry is their favorite journal and they greatly appreciate it due to the practical, useful, and pithy clinical updates it provides them as busy clinicians.

Current Psychiatry was born on January 1, 2002, and will be 21 years old at its premature demise on December 31, 2023 (This reminds me of the Billy Joel song “Only the Good Die Young”). The first Editor-in-Chief was Randolph Hillard, MD, who at the time was the psychiatry chair at the University of Cincinnati. I succeeded him as Editor-in-Chief in 2006 and will have served in that role for 17 years when Current Psychiatry is sunset. I have established 2 other research journals, Schizophrenia Research and Biomarkers in Neuropsychiatry, both of which are thriving. However, editing Current Psychiatry has been one of the most gratifying roles I have had in my career because Current Psychiatry promotes sound, evidence-based clinical practice to its 45,000 psychiatric clinician readers, who provide care for millions of psychiatric patients of all ages and DSM-5-TR diagnostic categories every day.

As the saying goes: All good things eventually come to an end. I am so grateful to have had the opportunity to collaborate with a wonderful, highly competent editorial staff, as well as with outstanding colleagues who served on the editorial board all those years. A special shout-out to Jeff Bauer, the publishing staff editor, with whom I worked so closely. I very much appreciated all the authors and peer reviewers who contributed timely clinical articles month after month and made Current Psychiatry such a valuable, evidence-based educational medium.

This has been a unique journey for all of us who strived to transform Current Psychiatry into a prominent, must-read clinical journal. This valedictory is both a fond farewell and a warm appreciation to you, our loyal readers. I hope that in the future we will reconnect and interact again in another meaningful way, advocating for the health and welfare of our psychiatric patients.

Article PDF
Author and Disclosure Information

Henry A. Nasrallah, MD, DLFAPA
Editor-in-Chief

Issue
Current Psychiatry - 22(12)
Publications
Topics
Page Number
9
Sections
Author and Disclosure Information

Henry A. Nasrallah, MD, DLFAPA
Editor-in-Chief

Author and Disclosure Information

Henry A. Nasrallah, MD, DLFAPA
Editor-in-Chief

Article PDF
Article PDF

All that’s bright must fade,
The brightest still the fleetest;
All that’s sweet was made
But to be lost when sweetest.
Thomas Moore

I sometimes hold it half a sin
To put in words the grief I feel;
For words, like Nature, half reveal
And half conceal the Soul within.
Alfred, Lord Tennyson, In Memoriam

Dear Readers,

I have sad news to share with you. This is the last issue of Current Psychiatry.

During my travels around the country over the past 2 decades, countless psychiatrists have told me that Current Psychiatry is their favorite journal and they greatly appreciate it due to the practical, useful, and pithy clinical updates it provides them as busy clinicians.

Current Psychiatry was born on January 1, 2002, and will be 21 years old at its premature demise on December 31, 2023 (This reminds me of the Billy Joel song “Only the Good Die Young”). The first Editor-in-Chief was Randolph Hillard, MD, who at the time was the psychiatry chair at the University of Cincinnati. I succeeded him as Editor-in-Chief in 2006 and will have served in that role for 17 years when Current Psychiatry is sunset. I have established 2 other research journals, Schizophrenia Research and Biomarkers in Neuropsychiatry, both of which are thriving. However, editing Current Psychiatry has been one of the most gratifying roles I have had in my career because Current Psychiatry promotes sound, evidence-based clinical practice to its 45,000 psychiatric clinician readers, who provide care for millions of psychiatric patients of all ages and DSM-5-TR diagnostic categories every day.

As the saying goes: All good things eventually come to an end. I am so grateful to have had the opportunity to collaborate with a wonderful, highly competent editorial staff, as well as with outstanding colleagues who served on the editorial board all those years. A special shout-out to Jeff Bauer, the publishing staff editor, with whom I worked so closely. I very much appreciated all the authors and peer reviewers who contributed timely clinical articles month after month and made Current Psychiatry such a valuable, evidence-based educational medium.

This has been a unique journey for all of us who strived to transform Current Psychiatry into a prominent, must-read clinical journal. This valedictory is both a fond farewell and a warm appreciation to you, our loyal readers. I hope that in the future we will reconnect and interact again in another meaningful way, advocating for the health and welfare of our psychiatric patients.

All that’s bright must fade,
The brightest still the fleetest;
All that’s sweet was made
But to be lost when sweetest.
Thomas Moore

I sometimes hold it half a sin
To put in words the grief I feel;
For words, like Nature, half reveal
And half conceal the Soul within.
Alfred, Lord Tennyson, In Memoriam

Dear Readers,

I have sad news to share with you. This is the last issue of Current Psychiatry.

During my travels around the country over the past 2 decades, countless psychiatrists have told me that Current Psychiatry is their favorite journal and they greatly appreciate it due to the practical, useful, and pithy clinical updates it provides them as busy clinicians.

Current Psychiatry was born on January 1, 2002, and will be 21 years old at its premature demise on December 31, 2023 (This reminds me of the Billy Joel song “Only the Good Die Young”). The first Editor-in-Chief was Randolph Hillard, MD, who at the time was the psychiatry chair at the University of Cincinnati. I succeeded him as Editor-in-Chief in 2006 and will have served in that role for 17 years when Current Psychiatry is sunset. I have established 2 other research journals, Schizophrenia Research and Biomarkers in Neuropsychiatry, both of which are thriving. However, editing Current Psychiatry has been one of the most gratifying roles I have had in my career because Current Psychiatry promotes sound, evidence-based clinical practice to its 45,000 psychiatric clinician readers, who provide care for millions of psychiatric patients of all ages and DSM-5-TR diagnostic categories every day.

As the saying goes: All good things eventually come to an end. I am so grateful to have had the opportunity to collaborate with a wonderful, highly competent editorial staff, as well as with outstanding colleagues who served on the editorial board all those years. A special shout-out to Jeff Bauer, the publishing staff editor, with whom I worked so closely. I very much appreciated all the authors and peer reviewers who contributed timely clinical articles month after month and made Current Psychiatry such a valuable, evidence-based educational medium.

This has been a unique journey for all of us who strived to transform Current Psychiatry into a prominent, must-read clinical journal. This valedictory is both a fond farewell and a warm appreciation to you, our loyal readers. I hope that in the future we will reconnect and interact again in another meaningful way, advocating for the health and welfare of our psychiatric patients.

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
9
Page Number
9
Publications
Publications
Topics
Article Type
Display Headline
Valedictory
Display Headline
Valedictory
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1223 From the Editor</fileName> <TBEID>0C02EBDD.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EBDD</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Valedictory</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231129T081813</firstPublished> <LastPublished>20231129T081813</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231129T081813</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Henry A. Nasrallah, MD, DLFAPA</byline> <bylineText/> <bylineFull>Henry A. Nasrallah, MD, DLFAPA</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>9</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>All that’s bright must fade,The brightest still the fleetest;All that’s sweet was madeBut to be lost when sweetest.</metaDescription> <articlePDF>299319</articlePDF> <teaserImage/> <title>Valedictory</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">70</term> </sections> <topics> <term canonical="true">278</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002651.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Valedictory</title> <deck/> </itemMeta> <itemContent> <p>All that’s bright must fade,<br/><br/>The brightest still the fleetest;<br/><br/>All that’s sweet was made<br/><br/>But to be lost when sweetest.</p> <p> <i>Thomas Moore</i> </p> <p>I sometimes hold it half a sin<br/><br/>To put in words the grief I feel;<br/><br/>For words, like Nature, half reveal<br/><br/>And half conceal the Soul within.</p> <p> <i>Alfred, Lord Tennyson, In Memoriam</i> </p> <p><br/><br/>Dear Readers,<br/><br/>I have sad news to share with you. This is the last issue of <scaps>Current Psychiatry</scaps>.</p> <p>During my travels around the country over the past 2 decades, countless psychiatrists have told me that <scaps>Current Psychiatry</scaps> is their favorite journal and they greatly appreciate it due to the practical, useful, and pithy clinical updates it provides them as busy clinicians.<br/><br/><scaps>Current Psychiatry</scaps> was born on January 1, 2002, and will be 21 years old at its premature demise on December 31, 2023 (This reminds me of the Billy Joel song “Only the Good Die Young”). The first Editor-in-Chief was Randolph Hillard, MD, who at the time was the psychiatry chair at the University of Cincinnati. I succeeded him as Editor-in-Chief in 2006 and will have served in that role for 17 years when <scaps>Current Psychiatry</scaps> is sunset. I have established 2 other research journals, <i>Schizophrenia Research</i> and <i>Biomarkers in Neuropsychiatry</i>, both of which are thriving. However, editing <scaps>Current Psychiatry</scaps> has been one of the most gratifying roles I have had in my career because <scaps>Current Psychiatry</scaps> promotes sound, evidence-based clinical practice to its 45,000 psychiatric clinician readers, who provide care for millions of psychiatric patients of all ages and DSM-5-TR diagnostic categories every day.<br/><br/>As the saying goes: All good things eventually come to an end. I am so grateful to have had the opportunity to collaborate with a wonderful, highly competent editorial staff, as well as with outstanding colleagues who served on the editorial board all those years. A special shout-out to Jeff Bauer, the publishing staff editor, with whom I worked so closely. I very much appreciated all the authors and peer reviewers who contributed timely clinical articles month after month and made <scaps>Current Psychiatry</scaps> such a valuable, evidence-based educational medium.<br/><br/>This has been a unique journey for all of us who strived to transform <scaps>Current Psychiatry</scaps> into a prominent, must-read clinical journal. This valedictory is both a fond farewell and a warm appreciation to you, our loyal readers. I hope that in the future we will reconnect and interact again in another meaningful way, advocating for the health and welfare of our psychiatric patients.</p> <p> <strong>Henry A. Nasrallah, MD, DLFAPA</strong> </p> <p>Editor-in-Chief</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002651.SIG
Disable zoom
Off

More on treating chronic insomnia

Article Type
Changed
Fri, 12/01/2023 - 01:15
Display Headline
More on treating chronic insomnia

In “Treating chronic insomnia: An alternating medication strategy” (Current Psychiatry, October 2023, p. 25-31, doi:10.12788/cp.0397), Dr. Kaplan correctly identified tolerance and tachyphylaxis as significant problems when prescribing traditional hypnotics, and proposed a solution of using 2 sleep medications, each having a different mechanism of action, on an alternating schedule. However, with the availability of the dual orexin receptor antagonists (DORAs) daridorexant, lemborexant, and suvorexant, this approach is unnecessary. Moreover, the mechanism of action of orexin receptor antagonism directly addresses extant hyperarousal by decreasing wake signaling, without any deleterious effect on sleep architecture.1 Additionally, the DORAs are not associated with physiological dependence, withdrawal, or rebound. Their efficacy profile is as good as or better than other FDA-approved agents for insomnia disorder.1 An obstacle to their use is that they are not yet available as generic products, but access is facilitated by the manufacturers’ patient assistance programs. Additional resources elaborate on indirect comparisons among agents using number needed to treat and number needed to harm, metrics that are helpful when clinically appraising new agents.2-5

Leslie Citrome, MD, MPH
Valhalla, New York

[embed:render:related:node:265588]

 

 

 

References

1. Rosenberg R, Citrome L, Drake CL. Advances in the treatment of chronic insomnia: a narrative review of new nonpharmacologic and pharmacologic therapies. Neuropsychiatr Dis Treat. 2021;17:2549-2566.

2. Citrome L. Dissecting clinical trials with ‘number needed to treat.’ Current Psychiatry. 2007;6(3):66-71.

3. Citrome L. Suvorexant for insomnia: a systematic review of the efficacy and safety profile for this newly approved hypnotic - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract. 2014;68(12):1429-1441.

4. Citrome L, Juday TR, Frech F, et al. Lemborexant for the treatment of insomnia: direct and indirect comparisons with other hypnotics using number needed to treat, number needed to harm, and likelihood to be helped or harmed. J Clin Psychiatry. 2021;82:20m13795. doi:10.4088/JCP.20m13795

5. Citrome L, Juday TR, Lundwall C. Lemborexant and daridorexant for the treatment of insomnia: an indirect comparison using number needed to treat, number needed to harm, and likelihood to be helped or harmed. J Clin Psychiatry. 2023;84(6):23m14851. doi:10.4088/JCP.23m14851

Article PDF
Author and Disclosure Information

Disclosures
Dr. Citrome is a consultant for AbbVie/Allergan, Acadia, Adamas, Alkermes, Angelini, Astellas, Avanir, Axsome, BioXcel, Boehringer Ingelheim, Cadent Therapeutics, Cerevel, Clinilabs, COMPASS, Eisai, Enteris BioPharma, HLS Therapeutics, Idorsia, Impel, INmune Bio, Intra-Cellular Therapies, Janssen, Karuna, Lundbeck, Lyndra, Medavante-ProPhase, Marvin, Merck, Mitsubishi-Tanabe Pharma, Neurelis, Neurocrine, Novartis, Noven, Otsuka, Ovid, Praxis, Recordati, Relmada, Reviva, Sage, Sunovion, Supernus, Teva, the University of Arizona, and Vanda, and has done one-off ad hoc consulting for individuals/entities conducting marketing, commercial, or scientific scoping research. He is a speaker for AbbVie/Allergan, Acadia, Alkermes, Angelini, Axsome, BioXcel, Eisai, Idorsia, Intra-Cellular Therapies, Janssen, Lundbeck, Neurocrine, Noven, Otsuka, Recordati, Sage, Sunovion, Takeda, Teva, and CME activities organized by medical education companies such as Medscape, NACCME, NEI, Vindico, universities, and professional organizations/societies.

Issue
Current Psychiatry - 22(12)
Publications
Topics
Page Number
10
Sections
Author and Disclosure Information

Disclosures
Dr. Citrome is a consultant for AbbVie/Allergan, Acadia, Adamas, Alkermes, Angelini, Astellas, Avanir, Axsome, BioXcel, Boehringer Ingelheim, Cadent Therapeutics, Cerevel, Clinilabs, COMPASS, Eisai, Enteris BioPharma, HLS Therapeutics, Idorsia, Impel, INmune Bio, Intra-Cellular Therapies, Janssen, Karuna, Lundbeck, Lyndra, Medavante-ProPhase, Marvin, Merck, Mitsubishi-Tanabe Pharma, Neurelis, Neurocrine, Novartis, Noven, Otsuka, Ovid, Praxis, Recordati, Relmada, Reviva, Sage, Sunovion, Supernus, Teva, the University of Arizona, and Vanda, and has done one-off ad hoc consulting for individuals/entities conducting marketing, commercial, or scientific scoping research. He is a speaker for AbbVie/Allergan, Acadia, Alkermes, Angelini, Axsome, BioXcel, Eisai, Idorsia, Intra-Cellular Therapies, Janssen, Lundbeck, Neurocrine, Noven, Otsuka, Recordati, Sage, Sunovion, Takeda, Teva, and CME activities organized by medical education companies such as Medscape, NACCME, NEI, Vindico, universities, and professional organizations/societies.

Author and Disclosure Information

Disclosures
Dr. Citrome is a consultant for AbbVie/Allergan, Acadia, Adamas, Alkermes, Angelini, Astellas, Avanir, Axsome, BioXcel, Boehringer Ingelheim, Cadent Therapeutics, Cerevel, Clinilabs, COMPASS, Eisai, Enteris BioPharma, HLS Therapeutics, Idorsia, Impel, INmune Bio, Intra-Cellular Therapies, Janssen, Karuna, Lundbeck, Lyndra, Medavante-ProPhase, Marvin, Merck, Mitsubishi-Tanabe Pharma, Neurelis, Neurocrine, Novartis, Noven, Otsuka, Ovid, Praxis, Recordati, Relmada, Reviva, Sage, Sunovion, Supernus, Teva, the University of Arizona, and Vanda, and has done one-off ad hoc consulting for individuals/entities conducting marketing, commercial, or scientific scoping research. He is a speaker for AbbVie/Allergan, Acadia, Alkermes, Angelini, Axsome, BioXcel, Eisai, Idorsia, Intra-Cellular Therapies, Janssen, Lundbeck, Neurocrine, Noven, Otsuka, Recordati, Sage, Sunovion, Takeda, Teva, and CME activities organized by medical education companies such as Medscape, NACCME, NEI, Vindico, universities, and professional organizations/societies.

Article PDF
Article PDF

In “Treating chronic insomnia: An alternating medication strategy” (Current Psychiatry, October 2023, p. 25-31, doi:10.12788/cp.0397), Dr. Kaplan correctly identified tolerance and tachyphylaxis as significant problems when prescribing traditional hypnotics, and proposed a solution of using 2 sleep medications, each having a different mechanism of action, on an alternating schedule. However, with the availability of the dual orexin receptor antagonists (DORAs) daridorexant, lemborexant, and suvorexant, this approach is unnecessary. Moreover, the mechanism of action of orexin receptor antagonism directly addresses extant hyperarousal by decreasing wake signaling, without any deleterious effect on sleep architecture.1 Additionally, the DORAs are not associated with physiological dependence, withdrawal, or rebound. Their efficacy profile is as good as or better than other FDA-approved agents for insomnia disorder.1 An obstacle to their use is that they are not yet available as generic products, but access is facilitated by the manufacturers’ patient assistance programs. Additional resources elaborate on indirect comparisons among agents using number needed to treat and number needed to harm, metrics that are helpful when clinically appraising new agents.2-5

Leslie Citrome, MD, MPH
Valhalla, New York

[embed:render:related:node:265588]

 

 

 

In “Treating chronic insomnia: An alternating medication strategy” (Current Psychiatry, October 2023, p. 25-31, doi:10.12788/cp.0397), Dr. Kaplan correctly identified tolerance and tachyphylaxis as significant problems when prescribing traditional hypnotics, and proposed a solution of using 2 sleep medications, each having a different mechanism of action, on an alternating schedule. However, with the availability of the dual orexin receptor antagonists (DORAs) daridorexant, lemborexant, and suvorexant, this approach is unnecessary. Moreover, the mechanism of action of orexin receptor antagonism directly addresses extant hyperarousal by decreasing wake signaling, without any deleterious effect on sleep architecture.1 Additionally, the DORAs are not associated with physiological dependence, withdrawal, or rebound. Their efficacy profile is as good as or better than other FDA-approved agents for insomnia disorder.1 An obstacle to their use is that they are not yet available as generic products, but access is facilitated by the manufacturers’ patient assistance programs. Additional resources elaborate on indirect comparisons among agents using number needed to treat and number needed to harm, metrics that are helpful when clinically appraising new agents.2-5

Leslie Citrome, MD, MPH
Valhalla, New York

[embed:render:related:node:265588]

 

 

 

References

1. Rosenberg R, Citrome L, Drake CL. Advances in the treatment of chronic insomnia: a narrative review of new nonpharmacologic and pharmacologic therapies. Neuropsychiatr Dis Treat. 2021;17:2549-2566.

2. Citrome L. Dissecting clinical trials with ‘number needed to treat.’ Current Psychiatry. 2007;6(3):66-71.

3. Citrome L. Suvorexant for insomnia: a systematic review of the efficacy and safety profile for this newly approved hypnotic - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract. 2014;68(12):1429-1441.

4. Citrome L, Juday TR, Frech F, et al. Lemborexant for the treatment of insomnia: direct and indirect comparisons with other hypnotics using number needed to treat, number needed to harm, and likelihood to be helped or harmed. J Clin Psychiatry. 2021;82:20m13795. doi:10.4088/JCP.20m13795

5. Citrome L, Juday TR, Lundwall C. Lemborexant and daridorexant for the treatment of insomnia: an indirect comparison using number needed to treat, number needed to harm, and likelihood to be helped or harmed. J Clin Psychiatry. 2023;84(6):23m14851. doi:10.4088/JCP.23m14851

References

1. Rosenberg R, Citrome L, Drake CL. Advances in the treatment of chronic insomnia: a narrative review of new nonpharmacologic and pharmacologic therapies. Neuropsychiatr Dis Treat. 2021;17:2549-2566.

2. Citrome L. Dissecting clinical trials with ‘number needed to treat.’ Current Psychiatry. 2007;6(3):66-71.

3. Citrome L. Suvorexant for insomnia: a systematic review of the efficacy and safety profile for this newly approved hypnotic - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract. 2014;68(12):1429-1441.

4. Citrome L, Juday TR, Frech F, et al. Lemborexant for the treatment of insomnia: direct and indirect comparisons with other hypnotics using number needed to treat, number needed to harm, and likelihood to be helped or harmed. J Clin Psychiatry. 2021;82:20m13795. doi:10.4088/JCP.20m13795

5. Citrome L, Juday TR, Lundwall C. Lemborexant and daridorexant for the treatment of insomnia: an indirect comparison using number needed to treat, number needed to harm, and likelihood to be helped or harmed. J Clin Psychiatry. 2023;84(6):23m14851. doi:10.4088/JCP.23m14851

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
10
Page Number
10
Publications
Publications
Topics
Article Type
Display Headline
More on treating chronic insomnia
Display Headline
More on treating chronic insomnia
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1223 Letters</fileName> <TBEID>0C02EA61.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EA61</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>More on treating chronic&#13;insomn</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231129T082050</firstPublished> <LastPublished>20231129T082050</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231129T082050</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Leslie Citrome, MD, MPH</byline> <bylineText/> <bylineFull>Leslie Citrome, MD, MPH</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>10</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>In “Treating chronic insomnia: An alternating medication strategy” (Current Psychiatry, October 2023, p. 25-31, doi:10.12788/cp.0397), Dr. Kaplan correctly iden</metaDescription> <articlePDF>299321</articlePDF> <teaserImage/> <title/> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords> <keyword>sleep medicine</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">52</term> </sections> <topics> <term canonical="true">296</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002652.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <h3>More on treating chronic insomnia</h3> <p>In “Treating chronic insomnia: An alternating medication strategy” (<scaps>Current Psychiatry</scaps>, October 2023, p. 25-31, doi:10.12788/cp.0397), Dr. Kaplan correctly identified tolerance and tachyphylaxis as significant problems when prescribing traditional hypnotics, and proposed a solution of using 2 sleep medications, each having a different mechanism of action, on an alternating schedule. However, with the availability of the dual orexin receptor antagonists (DORAs) daridorexant, lemborexant, and suvorexant, this approach is unnecessary. Moreover, the mechanism of action of orexin receptor antagonism directly addresses extant hyperarousal by decreasing wake signaling, without any deleterious effect on sleep architecture.<sup>1</sup> Additionally, the DORAs are not associated with physiological dependence, withdrawal, or rebound. Their efficacy profile is as good as or better than other FDA-approved agents for insomnia disorder.<sup>1</sup> An obstacle to their use is that they are not yet available as generic products, but access is facilitated by the manufacturers’ patient assistance programs. Additional resources elaborate on indirect comparisons among agents using number needed to treat and number needed to harm, metrics that are helpful when clinically appraising new agents.<sup>2-5</sup></p> <p> <strong>Leslie Citrome, MD, MPH </strong> </p> <p>Valhalla, New York</p> <p> <strong>References</strong> </p> <p class="reference"> 1. Rosenberg R, Citrome L, Drake CL. Advances in the treatment of chronic insomnia: a narrative review of new nonpharmacologic and pharmacologic therapies. Neuropsychiatr Dis Treat. 2021;17:2549-2566.<br/><br/> 2. Citrome L. Dissecting clinical trials with ‘number needed to treat.’ Current Psychiatry. 2007;6(3):66-71.<br/><br/> 3. Citrome L. Suvorexant for insomnia: a systematic review of the efficacy and safety profile for this newly approved hypnotic - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract. 2014;68(12):1429-1441.<br/><br/> 4. Citrome L, Juday TR, Frech F, et al. Lemborexant for the treatment of insomnia: direct and indirect comparisons with other hypnotics using number needed to treat, number needed to harm, and likelihood to be helped or harmed. J Clin Psychiatry. 2021;82:20m13795. doi:10.4088/JCP.20m13795<br/><br/> 5. Citrome L, Juday TR, Lundwall C. Lemborexant and daridorexant for the treatment of insomnia: an indirect comparison using number needed to treat, number needed to harm, and likelihood to be helped or harmed. J Clin Psychiatry. 2023;84(6):23m14851. doi:10.4088/JCP.23m14851</p> <p> <strong>Disclosures</strong> </p> <p class="disclosure">Dr. Citrome is a consultant for AbbVie/Allergan, Acadia, Adamas, Alkermes, Angelini, Astellas, Avanir, Axsome, BioXcel, Boehringer Ingelheim, Cadent Therapeutics, Cerevel, Clinilabs, COMPASS, Eisai, Enteris BioPharma, HLS Therapeutics, Idorsia, Impel, INmune Bio, Intra-Cellular Therapies, Janssen, Karuna, Lundbeck, Lyndra, Medavante-ProPhase, Marvin, Merck, Mitsubishi-Tanabe Pharma, Neurelis, Neurocrine, Novartis, Noven, Otsuka, Ovid, Praxis, Recordati, Relmada, Reviva, Sage, Sunovion, Supernus, Teva, the University of Arizona, and Vanda, and has done one-off ad hoc consulting for individuals/entities conducting marketing, commercial, or scientific scoping research. He is a speaker for AbbVie/Allergan, Acadia, Alkermes, Angelini, Axsome, BioXcel, Eisai, Idorsia, Intra-Cellular Therapies, Janssen, Lundbeck, Neurocrine, Noven, Otsuka, Recordati, Sage, Sunovion, Takeda, Teva, and CME activities organized by medical education companies such as Medscape, NACCME, NEI, Vindico, universities, and professional organizations/societies.</p> <p class="disclosure">doi: 10.12788/cp.0421</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002652.SIG
Disable zoom
Off

Monoamine oxidase inhibitors and tricyclic antidepressants for MDD

Article Type
Changed
Wed, 12/06/2023 - 10:08
Display Headline
Monoamine oxidase inhibitors and tricyclic antidepressants for MDD

CP02212035_pp.png

Ms. B, age 45, has a history of major depressive disorder (MDD) and migraines. She is admitted after presenting with anhedonia, hopelessness, and hypersomnia. These symptoms have become more severe over the last few weeks. Ms. B describes a past suicide attempt via overdose on doxylamine for which she required treatment in the intensive care unit. The only activity she enjoys is her weekly girls’ night, during which she drinks a few glasses of wine. Ms. B’s current medications are dextromethorphan/bupropion 45/105 mg twice daily and aripiprazole 5 mg/d, which she has taken for 3 months. She states she has “been on every antidepressant there is.”

When clinicians review Ms. B’s medication history, it is clear she has had adequate trials of selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), intranasal esketamine, multiple augmentation strategies, and electroconvulsive therapy (ECT). Ms. B seeks an alternative medication to improve her depressive symptoms.

Treatment-resistant depression (TRD) is commonly defined as depression that has not responded to ≥2 adequate trials of an antidepressant.1 Some guidelines recommend monoamine oxidase inhibitors (MAOIs) and tricyclic antidepressants (TCAs) as second- or even third-line options for MDD,2 while others recommend reserving them for patients with insufficient responses to alternative treatment modalities.3,4 Although MAOIs and TCAs have been available since the 1950s, prescribing these medications has become less prevalent due to safety concerns, the availability of other pharmacologic options, and a lack of clinical training and comfort.5,6 Most research notes that MAOIs are superior for treating atypical depression while TCAs are more effective for melancholic depression.2-4 In a review of 20 studies, Thase et al7 found that 50% of TCA nonresponders benefited from an MAOI. In the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) trial, monotherapy with the MAOI tranylcypromine was associated with a lower remission rate than the TCA nortriptyline; many argue the dose of tranylcypromine was suboptimal, and few participants completed an adequate trial in the last level.8,9 A more recent study by Kim et al10 found MAOIs to be “generally more effective” than TCAs for TRD, particularly in patients with fewer antidepressant trials; however, this was a small retrospective exploratory trial. A network meta-analysis found both classes to be “competitive” with SSRIs based on efficacy and tolerability, which leads to the question of whether these medications should be considered earlier in therapy.11 Considering patient-specific factors and particular medication properties is an effective strategy when prescribing an MAOI or TCA.

Monoamine oxidase inhibitors

Four MAOIs are FDA-approved for treating MDD (Table 15,12-17): phenelzine, isocarboxazid, tranylcypromine, and selegiline. These medications irreversibly inhibit MAO, which exists as isomers A and B. MAO-A primarily metabolizes serotonin and norepinephrine, which is largely responsible for these medications’ antidepressant effects. Both isomers equally metabolize dopamine.5,12,18 It is best to avoid using MAOIs in patients with cerebrovascular disease, hepatic disease, or pheochromocytoma. Patients with active substance use disorders (particularly sympathomimetics and hallucinogens) are at an increased risk for hypertensive crises and serotonin syndrome, respectively. The most common adverse effects are orthostatic hypotension (despite more well-known concerns regarding hypertension), alterations in sleep patterns (insomnia or hypersomnia, depending on the agent), gastrointestinal issues, and anticholinergic adverse effects such as dry mouth and constipation.13,19-21

CP02212035_t1.png

In one review and meta-analysis, phenelzine displayed the highest efficacy across all MAOIs.11 It likely requires high doses to achieve adequate MAO inhibition.11 A metabolite of phenelzine inhibits gamma-aminobutyric acid transaminase and may be helpful for patients with comorbid anxiety disorders or MDD with anxious distress.18,21 Additional considerations include phenelzine’s propensity for orthostasis (with rapid titrations and higher doses), sedation, weight gain, sexual dysfunction, and a rare adverse effect of vitamin B6 deficiency.5,13,14,20-22

Use of isocarboxazid in clinical practice is rare. Its adverse effects are similar to those of phenelzine but isocarboxazid is less studied. Tranylcypromine has a similar chemical structure to amphetamine. It can be stimulating at higher doses, potentially benefitting patients with comorbid attention-deficit/hyperactivity disorder (ADHD) or significant apathy.13,23 Selegiline’s distinct quality is its availability as a transdermal patch, which may be useful for patients who struggle to take oral medications. At low doses (6 mg/24 h), the selegiline transdermal patch allows patients to disregard a dietary tyramine restriction because it avoids first-pass metabolism. It inhibits both MAO isomers in the brain but is only selective for MAO-B once concentrations are distributed to the liver. Higher doses require a tyramine-restricted diet because there is still some MAO-A inhibition in the gut. Selegiline is also stimulating because it is converted to amphetamine and methamphetamine.5,12,13,17,19,24

Despite promising results from the use of MAOIs, physicians and patients may be reluctant to use these medications due to perceived limitations. One prominent barrier is the infamous “cheese reaction.” Tyramine, an amino acid found in certain food and beverages (Table 25,13-18,25-28), is broken down by MAO-A in the gut. When this enzyme is inhibited, higher concentrations of tyramine reach systemic circulation. Tyramine’s release of norepinephrine (which now cannot be broken down) can lead to a hypertensive crisis. Consequently, a tyramine-restricted diet is recommended for patients taking an MAOI. However, the common notion that cheese, wine, and beer must be avoided is false, because most of the dietary restrictions developed following the discovery of MAOIs are antiquated.5,12,25-28 Patients who take an MAOI only need to slightly adjust their diet, as outlined in Table 2.5,13-18,25-28 A reasonable serving size of most foods and beverages containing tyramine is unlikely to elicit this “pressor” response. Of the 4 MAOIs FDA-approved for MDD, tranylcypromine appears to be the most sensitive to tyramine.21 Transient postdose hypertension (regardless of tyramine) may occur after taking an MAOI.29 Encourage patients to monitor their blood pressure.

CP02212035_t2.png

Continue to: Additional hurdles include...

 

 

Additional hurdles include the required washout period from serotonergic medications and interactions with sympathomimetics. MAOIs pose the highest risk of serotonin syndrome; however, this usually occurs if given concomitantly with other serotonergic agents. The standard recommendation is a 14-day washout period from SSRIs (5 weeks for fluoxetine and 3 weeks for vortioxetine), SNRIs, mirtazapine, and other antidepressants. It can be distressing for patients to be without medication during that period. Because some antidepressants have much shorter half-lives, waiting 5 half-lives (typically 5 to 7 days) for the discontinued medication to be excreted is feasible if patients are closely monitored.5,12,13,25,27,30 There are rare instances where a TCA may be combined with an MAOI (typically initiated within 1 to 2 days of each other), but never clomipramine or imipramine due to their potent serotonin reuptake inhibition.31 If switching to an alternative MAOI, waiting 7 to 14 days is recommended to allow adequate time for the inhibited enzyme to regenerate.14-17,32 Taking medications that increase dopamine and norepinephrine (eg, stimulants or oral over-the-counter decongestants) with an MAOI is typically not recommended due to the risk of hypertensive crisis.25,27 In severe TRD or comorbid ADHD, successful simultaneous use of methylphenidate or amphetamine—typically at low doses—with close blood pressure monitoring has been reported.33 There have also been positive cases of the use of modafinil in combination with an MAOI; however, this should be done with caution.34,35 Clinicians must use clinical judgment when considering a combination of medications that pose a higher risk.

Tricyclic antidepressants

TCAs work differently than MAOIs to increase monoamines. They inhibit presynaptic serotonin and norepinephrine transporters in the CNS to increase levels of these chemicals in the synaptic cleft. While all TCAs inhibit these transporters, they do so at varying levels (Table 336-51). Based on their chemical structure, TCAs can be categorized into secondary and tertiary amines. Tertiary amines are metabolized via demethylation into their derivatives (Table 336-51). Patients who have recently suffered a myocardial infarction (MI) should avoid tertiary amines. TCAs can reduce heart rate variability, which is already decreased after an MI, thus presenting the potential for cardiac arrhythmias. TCAs should also be avoided in patients with cardiac conduction abnormalities.38-46,52 Patients with a prior baseline cardiac conduction defect, such as a bundle branch block, are at higher risk for further cardiac abnormalities. In those with a preexisting first-degree heart block, TCAs can still be used, but electrocardiogram monitoring is recommended.52,53 TCAs have also been reported to decrease the seizure threshold.38-46 They can be used with caution in patients who have a history of epilepsy or head trauma, or with concomitant medications that lower the seizure threshold.38-46

CP02212035_t3.png

Overdose risk is a concern with TCAs because ingestion of 10 to 20 mg/kg can lead to significant toxicity.54 This is due to their blockage of voltage-gated sodium channels found in the CNS and heart, which contributes to overdose symptoms such as a widened QRS complex and seizures. Symptoms usually develop within 2 hours but may be delayed up to 6 hours.55 Patients with a history of overdose must be carefully assessed before initiating a TCA. Prescribing a limited supply of these medications may be valuable. The use of TCAs has often been limited due to their adverse effects, most of which are associated with their respective affinities for alpha 1, muscarinic 1, and histamine 1 receptors. Inhibition of the alpha 1 receptor is associated with hypotension, muscarinic 1 with anticholinergic adverse effects, and histamine 1 with sedation and weight gain. Tertiary amines have a higher affinity for these receptors compared to secondary amines, leading to a more significant adverse effect profile.36,50 Among TCAs, amitriptyline is the most likely to cause hypotension, whereas desipramine and nortriptyline are least likely. Amitriptyline and clomipramine are most likely to cause anticholinergic adverse effects, whereas desipramine and nortriptyline are the least likely. Amitriptyline, doxepin, and imipramine have the highest propensity for QTc prolongation.36

Beyond treating MDD, TCAs have shown benefits for treating other disease states (Table 438-46,49,56-61).These differing indications may help psychiatrists determine the best TCA to prescribe for a given patient. Amitriptyline is the most studied TCA for MDD; however, nortriptyline is typically preferred due to its favorable tolerability profile.4,62 Nortriptyline also has data supporting its use in ECT to prevent relapse.63 Amitriptyline and nortriptyline have shown benefits in patients with neuro­pathic pain and for migraine prophylaxis.56-60 Although frequently used for MDD, clomipramine is not FDA-approved for this indication, but is for obsessive-compulsive disorder.39 Doxepin is FDA-approved for insomnia at lower doses and for MDD at higher doses.40 Therefore, it may benefit patients with sleep difficulties secondary to depression. Desipramine has been used off-label to treat ADHD in children and has shown some benefits in adults.64-66 Protriptyline, trimipramine, and amoxapine are infrequently used in clinical practice.

CP02212035_t4.png

A unique feature of TCAs is the ability to monitor serum concentrations (Table 336-51).Guidelines recommend therapeutic drug monitoring (TDM) with amitriptyline, clomi­pramine, imipramine, and nortriptyline for routine use. TDM is still recommended for doxepin, desipramine, and trimipramine, but its utility is largely for treatment failure or resistance.37 These plasma levels can be altered based on coadministered medications (Table 538-46) and should be closely monitored. Physicians should obtain a trough level after at least 5 half-lives and before the next dose is due, and use TDM as indicated to optimize dosing.

CP02212035_t5.png

Continue to: CASE CONTINUED

 

 

CASE CONTINUED

Ms. B’s outpatient psychiatrist provides collateral information about her medical history and confirms her long-standing MDD with multiple medication trials, though she has never received an MAOI or TCA. Ms. B is adamant she does not want a medication-free period between treatments and refuses to adjust her diet, despite being educated on the few changes necessary. She has no contraindications for TCAs and may benefit from a TCA for her comorbid migraines. The care team expresses concern for TCA overdose to Ms. B and her family. Ms. B’s sister reassures the team they will have someone monitor and dispense her medications at home. They decide to discontinue her current psychiatric regimen, and Ms. B is started on nortriptyline 50 mg/d at night, with plans to titrate based on tolerability.

Related Resources

Drug Brand Names

Amitriptyline • Elavil
Amphetamine • Adzenys, Dyanavel
Aripiprazole • Abilify
Clomipramine • Anafranil
Desipramine • Norpramin
Dextromethorphan/bupropion • Auvelity
Doxepin • Sinequan, Adapin
Esketamine • Spravato
Fluoxetine • Prozac
Imipramine • Tofranil
Isocarboxazid • Marplan
Methamphetamine • Desoxyn
Mirtazapine • Remeron
Modafinil • Provigil
Nortriptyline • Pamelor
Phenelzine • Nardil
Protriptyline • Vivactil
Selegiline • Emsam
Tranylcypromine • Parnate
Trimipramine • Surmontil
Vortioxetine • Trintellix

[embed:render:related:node:153015]

[embed:render:related:node:246706]

References

1. Gaynes BN, Lux L, Gartlehner G, et al. Defining treatment-resistant depression. Depress Anxiety. 2020;37(2):134-145. doi:10.1002/da.22968

2. Kennedy SH, Lam RW, McIntyre RS, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 clinical guidelines for the management of adults with major depressive disorder: section 3. Pharmacological treatments. Can J Psychiatry. 2016;61(9):540-560. doi:10.1177/0706743716659417

3. VA/DoD clinical practice guideline for the management of major depressive disorder. Veterans Health Administration and Department of Defense; 2016. https://www.healthquality.va.gov/guidelines/MH/mdd/VADoDMDDCPGFINAL82916.pdf

4. Gelenberg AJ, Freeman MP, Markowitz JC, et al. Practice guideline for the treatment of patients with major depressive disorder. Am J Psychiatry. 2010;167(Suppl 10):9-118.

5. Meyer JM. A concise guide to monoamine oxidase inhibitors. Current Psychiatry. 2017;16(12):14-16,18-23,47,A.

6. Taylor D. Selective serotonin reuptake inhibitors and tricyclic antidepressants in combination. Interactions and therapeutic uses. Br J Psychiatry. 1995;167(5):575-580. doi:10.1192/bjp.167.5.575

7. Thase ME, Trivedi MH, Rush AJ. MAOIs in the contemporary treatment of depression. Neuropsychopharmacology. 1995;12(3):185-219. doi:10.1016/0893-133X(94)00058-8

8. Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006;163(11):1905-1917. doi:10.1176/ajp.2006.163.11.1905

9. McGrath PJ, Stewart JW, Fava M, et al. Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: a STAR*D report. Am J Psychiatry. 2006;163(9):1531-1666. doi:10.1176/ajp.2006.163.9.1531

10. Kim T, Xu C, Amsterdam JD. Relative effectiveness of tricyclic antidepressant versus monoamine oxidase inhibitor monotherapy for treatment-resistant depression. J Affect Disord. 2019;250:199-203. doi:10.1016/j.jad.2019.03.028

11. Suchting R, Tirumalajaru V, Gareeb R, et al. Revisiting monoamine oxidase inhibitors for the treatment of depressive disorders: a systematic review and network meta-analysis. J Affect Disord. 2021;282:1153-1160. doi:10.1016/j.jad.2021.01.021


12. Stahl SM, Felker A. Monoamine oxidase inhibitors: a modern guide to an unrequited class of antidepressants. CNS Spectr. 2008;13(10):855-870. doi:10.1017/s1092852900016965

13. Chamberlain SR, Baldwin DS. Monoamine oxidase inhibitors (MAOIs) in psychiatric practice: how to use them safely and effectively. CNS Drugs. 2021;35(7):703-716. doi:10.1007/s40263-021-00832-x

14. Nardil [package insert]. New York, NY: Parke-Davis; 2009.

15. Marplan [package insert]. Parsippany, NJ: Validus Pharmaceuticals LLC; 2020.

16. Parnate [package insert]. Saint Michael, Barbados: Concordia Pharmaceuticals; 2015.

17. Emsam [package insert]. Morgantown, WV: Mylan Specialty LP; 2014.

18. Shulman KI, Herrmann N, Walker SE. Current place of monoamine oxidase inhibitors in the treatment of depression. CNS Drugs. 2013;27(10):789-797. doi:10.1007/s40263-013-0097-3

19. Sub Laban T, Saadabadi A. Monoamine oxidase inhibitors (MAOI). StatPearls Publishing; 2023. https://www.ncbi.nlm.nih.gov/books/NBK539848/

20. Rabkin JG, Quitkin FM, McGrath P, et al. Adverse reactions to monoamine oxidase inhibitors. Part II. Treatment correlates and clinical management. J Clin Psychopharmacol. 1985;5(1):2-9.

21. Gillman PK. Advances pertaining to the pharmacology and interactions of irreversible nonselective monoamine oxidase inhibitors. J Clin Psychopharmacol. 2011;31(1):66-74. doi:10.1097/JCP.0b013e31820469ea

22. Sidhu G, Marwaha R. Phenelzine. StatPearls Publishing; 2023. https://www.ncbi.nlm.nih.gov/books/NBK554508/

23. Frieling H, Bleich S. Tranylcypromine: new perspectives on an “old” drug. Eur Arch Psychiatry Clin Neurosci. 2006;256(5):268-273. doi:10.1007/s00406-006-0660-8

24. Goodnick PJ. Seligiline transdermal system in depression. Expert Opin Pharmacother. 2007;8(1):59-64. doi:10.1517/14656566.8.1.59

25. Edinoff AN, Swinford CR, Odisho AS, et al. Clinically relevant drug interactions with monoamine oxidase inhibitors. Health Psychol Res. 2022;10(4):39576. doi:10.52965/001c.39576

26. Gillman PK. A reassessment of the safety profile of monoamine oxidase inhibitors: elucidating tired old tyramine myths. J Neural Transm (Vienna). 2018;125(11):1707-1717. doi:10.1007/s00702-018-1932-y

27. Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03

28. McCabe-Sellers BJ, Staggs CG, Bogle ML. Tyramine in foods and monoamine oxidase inhibitor drugs: a crossroad where medicine, nutrition, pharmacy, and food industry converge. J Food Composit Anal. 2006;19:S58-S65. doi:10.1016/j.jfca.2005.12.008

29. Keck PE Jr, Vuckovic A, Pope HG Jr, et al. Acute cardiovascular response to monoamine oxidase inhibitors: a prospective assessment. J Clin Psychopharmacol. 1989;9(3):203-206.

30. Bodkin JA, Dunlop BW. Moving on with monoamine oxidase inhibitors. Focus (Am Psychiatr Publ). 2021;19(1):50-52. doi:10.1176/appi.focus.20200046

31. Amsterdam JD, Kim TT. Relative effectiveness of monoamine oxidase inhibitor and tricyclic antidepressant combination therapy for treatment-resistant depression. J Clin Psychopharmacol. 2019;39(6):649-652. doi:10.1097/JCP.0000000000001130

32. Keks N, Hope J, Keogh S. Switching and stopping antidepressants. Aust Prescr. 2016;39(3):76-83. doi:10.18773/austprescr.2016.039

33. Israel JA. Combining stimulants and monoamine oxidase inhibitors: a reexamination of the literature and a report of a new treatment combination. Prim Care Companion CNS Disord. 2015;17(6):10.4088/PCC.15br01836. doi:10.4088/PCC.15br01836

34. Clemons WE, Makela E, Young J. Concomitant use of modafinil and tranylcypromine in a patient with narcolepsy: a case report. Sleep Med. 2004;5(5):509-511. doi:10.1016/j.sleep.2004.06.006

35. Ashton AK. Modafinil augmentation of phenelzine for residual fatigue in dysthymia. Am J Psychiatry. 2004;161(9):1716-1717. doi:10.1176/appi.ajp.161.9.1716-a

36. O’Donnell JM, Bies RR, Shelton RC. Drug therapy of depression and anxiety disorders. In: Brunton LL, Hilal-Dandan R, Knollmann BC, eds. Goodman & Gilman’s: The Pharmacological Basis of Therapeutics. 13th ed. McGraw Hill; 2017. Accessed June 4, 2023. https://accessanesthesiology.mhmedical.com/content.aspx?bookid=2189&sectionid=169518711

37. Hiemke C, Bergemann N, Clement HW, et al. Consensus guidelines for therapeutic drug monitoring in neuropsychopharmacology: update 2017. Pharmacopsychiatry. 2018;51(1-02):9-62. doi:10.1055/s-0043-116492

38. Amitriptyline hydrochloride [package insert]. East Brunswick, NJ: Unichem Pharmaceuticals (USA); 2021.

39. Clomipramine hydrochloride [package insert]. East Windsor, NJ: Aurobindo Pharma Limited; 2023.

40. Doxepin hydrochloride capsules, USP [package insert]. Bedminster, NJ: Alembic Pharmaceuticals Inc; 2021. 

41. Imipramine hydrochloride tablet [package insert]. Fairfield, NJ: Leading Pharma LLC USA; 2022.  

42. Trimipramine maleate [package insert]. Northvale, NJ: Elite Laboratories Inc; 2021. 

43. Amoxapine [package insert]. Parsippany, NJ: Actavis Pharma Inc; 2015.

44. Desipramine hydrochloride tablets [package insert]. Bedminster, NJ: Alembic Pharmaceuticals Inc; 2023.

45. Nortriptyline hydrochloride capsules, USP [package insert]. Parsippany, NJ: Teva Pharmaceuticals Inc; 2021.

46. Protriptyline hydrochloride [package insert]. Bensalem, PA: Sigmapharm Laboratories, LLC; 2023.

47. Calvo B, García MJ, Pedraz JL, et al. Pharmacokinetics of amoxapine and its active metabolites. Int J Clin Pharmacol Ther Toxicol. 1985;23(4):180-185.

48. Ziegler VE, Biggs JT, Wylie LT, et al. Protriptyline kinetics. Clin Pharmacol Ther. 1978;23(5):580-584. doi:10.1002/cpt1978235580

49. Cleare A, Pariante CM, Young AH, et al. Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol. 2015;29(5):459-525. doi:10.1177/0269881115581093

50. Richelson E. Synaptic effects of antidepressants. J Clin Psychopharmacol. 1996;16(3 Suppl 2):1S-9S. doi:10.1097/00004714-199606002-00001

51. Vos CF, Aarnoutse RE, Op de Coul MJM, et al. Tricyclic antidepressants for major depressive disorder: a comprehensive evaluation of current practice in the Netherlands. BMC Psychiatry. 2021;21(1):481. doi:10.1186/s12888-021-03490-x

52. Alvarez W Jr, Pickworth KK. Safety of antidepressant drugs in the patient with cardiac disease: a review of the literature. Pharmacotherapy. 2003;23(6):754-771. doi:10.1592/phco.23.6.754.32185

53. Dietch JT, Fine M. The effect of nortriptyline in elderly patients with cardiac conduction disease. J Clin Psychiatry. 1990;51(2):65-67.

54. Valento M, Liebelt EL. Cyclic antidepressants. In: Nelson LS, Howland M, Lewin NA, et al, eds. Goldfrank’s Toxicologic Emergencies. 9th ed. McGraw Hill; 2011. Accessed June 10, 2023. https://accesspharmacy.mhmedical.com/content.aspx?bookid=2569&sectionid=210274664

55. Woolf AD, Erdman AR, Nelson LS, et al. Tricyclic antidepressant poisoning: an evidence-based consensus guideline for out-of-hospital management. Clin Toxicol (Phila). 2007;45(3):203-233. doi:10.1080/15563650701226192

56. Baldwin DS, Anderson IM, Nutt DJ, et al. Evidence-based pharmacological treatment of anxiety disorders, post-traumatic stress disorder and obsessive-compulsive disorder: a revision of the 2005 guidelines from the British Association for Psychopharmacology. J Psychopharmacol. 2014;28(5):403-439. doi:10.1177/0269881114525674

57. Moulin DE, Clark AJ, Gilron I, et al. Pharmacological management of chronic neuropathic pain - consensus statement and guidelines from the Canadian Pain Society. Pain Res Manag. 2007;12(1):13-21. doi:10.1155/2007/730785

58. Finnerup NB, Attal N, Haroutounian S, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015;14(2):162-173. doi:10.1016/S1474-4422(14)70251-0

59. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev. 2007;(4):CD005454. doi:10.1002/14651858.CD005454.pub2

60. Burch R. Antidepressants for preventive treatment of migraine. Curr Treat Options Neurol. 2019;21(4):18. doi:10.1007/s11940-019-0557-2

61. Ailani J, Burch RC, Robbins MS; Board of Directors of the American Headache Society. The American Headache Society consensus statement: update on integrating new migraine treatments into clinical practice. Headache. 2021;61(7):1021-1039. doi:10.1111/head.14153

62. Bauer M, Pfennig A, Severus E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for biological treatment of unipolar depressive disorders, part 1: update 2013 on the acute and continuation treatment of unipolar depressive disorders. World J Biol Psychiatry. 2013;14(5):334-385. doi:10.3109/15622975.2013.804195

63. Jelovac A, Kolshus E, McLoughlin DM. Relapse following successful electroconvulsive therapy for major depression: a meta-analysis. Neuropsychopharmacology. 2013;38(12):2467-2474. doi:10.1038/npp.2013.149

64. Spencer T, Biederman J, Coffey B, et al. A double-blind comparison of desipramine and placebo in children and adolescents with chronic tic disorder and comorbid attention-deficit/hyperactivity disorder. Arch Gen Psychiatry. 2002;59(7):649-656. doi:10.1001/archpsyc.59.7.649

65. Spencer T, Biederman J, Wilens T, et al. Pharmacotherapy of attention-deficit hyperactivity disorder across the life cycle. J Am Acad Child Adolesc Psychiatry. 1996;35(4):409-432. doi:10.1097/00004583-199604000-00008

66. Wilens TE, Biederman J, Prince J, et al. Six-week, double-blind, placebo-controlled study of desipramine for adult attention deficit hyperactivity disorder. Am J Psychiatry. 1996;153(9):1147-1153. doi:10.1176/ajp.153.9.1147

Article PDF
Author and Disclosure Information

Dr. Costello and Dr. Gleisner are Clinical Pharmacists, McLean Hospital, Belmont, Massachusetts.

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Issue
Current Psychiatry - 22(12)
Publications
Topics
Page Number
35-42
Sections
Author and Disclosure Information

Dr. Costello and Dr. Gleisner are Clinical Pharmacists, McLean Hospital, Belmont, Massachusetts.

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Author and Disclosure Information

Dr. Costello and Dr. Gleisner are Clinical Pharmacists, McLean Hospital, Belmont, Massachusetts.

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Article PDF
Article PDF

CP02212035_pp.png

Ms. B, age 45, has a history of major depressive disorder (MDD) and migraines. She is admitted after presenting with anhedonia, hopelessness, and hypersomnia. These symptoms have become more severe over the last few weeks. Ms. B describes a past suicide attempt via overdose on doxylamine for which she required treatment in the intensive care unit. The only activity she enjoys is her weekly girls’ night, during which she drinks a few glasses of wine. Ms. B’s current medications are dextromethorphan/bupropion 45/105 mg twice daily and aripiprazole 5 mg/d, which she has taken for 3 months. She states she has “been on every antidepressant there is.”

When clinicians review Ms. B’s medication history, it is clear she has had adequate trials of selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), intranasal esketamine, multiple augmentation strategies, and electroconvulsive therapy (ECT). Ms. B seeks an alternative medication to improve her depressive symptoms.

Treatment-resistant depression (TRD) is commonly defined as depression that has not responded to ≥2 adequate trials of an antidepressant.1 Some guidelines recommend monoamine oxidase inhibitors (MAOIs) and tricyclic antidepressants (TCAs) as second- or even third-line options for MDD,2 while others recommend reserving them for patients with insufficient responses to alternative treatment modalities.3,4 Although MAOIs and TCAs have been available since the 1950s, prescribing these medications has become less prevalent due to safety concerns, the availability of other pharmacologic options, and a lack of clinical training and comfort.5,6 Most research notes that MAOIs are superior for treating atypical depression while TCAs are more effective for melancholic depression.2-4 In a review of 20 studies, Thase et al7 found that 50% of TCA nonresponders benefited from an MAOI. In the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) trial, monotherapy with the MAOI tranylcypromine was associated with a lower remission rate than the TCA nortriptyline; many argue the dose of tranylcypromine was suboptimal, and few participants completed an adequate trial in the last level.8,9 A more recent study by Kim et al10 found MAOIs to be “generally more effective” than TCAs for TRD, particularly in patients with fewer antidepressant trials; however, this was a small retrospective exploratory trial. A network meta-analysis found both classes to be “competitive” with SSRIs based on efficacy and tolerability, which leads to the question of whether these medications should be considered earlier in therapy.11 Considering patient-specific factors and particular medication properties is an effective strategy when prescribing an MAOI or TCA.

Monoamine oxidase inhibitors

Four MAOIs are FDA-approved for treating MDD (Table 15,12-17): phenelzine, isocarboxazid, tranylcypromine, and selegiline. These medications irreversibly inhibit MAO, which exists as isomers A and B. MAO-A primarily metabolizes serotonin and norepinephrine, which is largely responsible for these medications’ antidepressant effects. Both isomers equally metabolize dopamine.5,12,18 It is best to avoid using MAOIs in patients with cerebrovascular disease, hepatic disease, or pheochromocytoma. Patients with active substance use disorders (particularly sympathomimetics and hallucinogens) are at an increased risk for hypertensive crises and serotonin syndrome, respectively. The most common adverse effects are orthostatic hypotension (despite more well-known concerns regarding hypertension), alterations in sleep patterns (insomnia or hypersomnia, depending on the agent), gastrointestinal issues, and anticholinergic adverse effects such as dry mouth and constipation.13,19-21

CP02212035_t1.png

In one review and meta-analysis, phenelzine displayed the highest efficacy across all MAOIs.11 It likely requires high doses to achieve adequate MAO inhibition.11 A metabolite of phenelzine inhibits gamma-aminobutyric acid transaminase and may be helpful for patients with comorbid anxiety disorders or MDD with anxious distress.18,21 Additional considerations include phenelzine’s propensity for orthostasis (with rapid titrations and higher doses), sedation, weight gain, sexual dysfunction, and a rare adverse effect of vitamin B6 deficiency.5,13,14,20-22

Use of isocarboxazid in clinical practice is rare. Its adverse effects are similar to those of phenelzine but isocarboxazid is less studied. Tranylcypromine has a similar chemical structure to amphetamine. It can be stimulating at higher doses, potentially benefitting patients with comorbid attention-deficit/hyperactivity disorder (ADHD) or significant apathy.13,23 Selegiline’s distinct quality is its availability as a transdermal patch, which may be useful for patients who struggle to take oral medications. At low doses (6 mg/24 h), the selegiline transdermal patch allows patients to disregard a dietary tyramine restriction because it avoids first-pass metabolism. It inhibits both MAO isomers in the brain but is only selective for MAO-B once concentrations are distributed to the liver. Higher doses require a tyramine-restricted diet because there is still some MAO-A inhibition in the gut. Selegiline is also stimulating because it is converted to amphetamine and methamphetamine.5,12,13,17,19,24

Despite promising results from the use of MAOIs, physicians and patients may be reluctant to use these medications due to perceived limitations. One prominent barrier is the infamous “cheese reaction.” Tyramine, an amino acid found in certain food and beverages (Table 25,13-18,25-28), is broken down by MAO-A in the gut. When this enzyme is inhibited, higher concentrations of tyramine reach systemic circulation. Tyramine’s release of norepinephrine (which now cannot be broken down) can lead to a hypertensive crisis. Consequently, a tyramine-restricted diet is recommended for patients taking an MAOI. However, the common notion that cheese, wine, and beer must be avoided is false, because most of the dietary restrictions developed following the discovery of MAOIs are antiquated.5,12,25-28 Patients who take an MAOI only need to slightly adjust their diet, as outlined in Table 2.5,13-18,25-28 A reasonable serving size of most foods and beverages containing tyramine is unlikely to elicit this “pressor” response. Of the 4 MAOIs FDA-approved for MDD, tranylcypromine appears to be the most sensitive to tyramine.21 Transient postdose hypertension (regardless of tyramine) may occur after taking an MAOI.29 Encourage patients to monitor their blood pressure.

CP02212035_t2.png

Continue to: Additional hurdles include...

 

 

Additional hurdles include the required washout period from serotonergic medications and interactions with sympathomimetics. MAOIs pose the highest risk of serotonin syndrome; however, this usually occurs if given concomitantly with other serotonergic agents. The standard recommendation is a 14-day washout period from SSRIs (5 weeks for fluoxetine and 3 weeks for vortioxetine), SNRIs, mirtazapine, and other antidepressants. It can be distressing for patients to be without medication during that period. Because some antidepressants have much shorter half-lives, waiting 5 half-lives (typically 5 to 7 days) for the discontinued medication to be excreted is feasible if patients are closely monitored.5,12,13,25,27,30 There are rare instances where a TCA may be combined with an MAOI (typically initiated within 1 to 2 days of each other), but never clomipramine or imipramine due to their potent serotonin reuptake inhibition.31 If switching to an alternative MAOI, waiting 7 to 14 days is recommended to allow adequate time for the inhibited enzyme to regenerate.14-17,32 Taking medications that increase dopamine and norepinephrine (eg, stimulants or oral over-the-counter decongestants) with an MAOI is typically not recommended due to the risk of hypertensive crisis.25,27 In severe TRD or comorbid ADHD, successful simultaneous use of methylphenidate or amphetamine—typically at low doses—with close blood pressure monitoring has been reported.33 There have also been positive cases of the use of modafinil in combination with an MAOI; however, this should be done with caution.34,35 Clinicians must use clinical judgment when considering a combination of medications that pose a higher risk.

Tricyclic antidepressants

TCAs work differently than MAOIs to increase monoamines. They inhibit presynaptic serotonin and norepinephrine transporters in the CNS to increase levels of these chemicals in the synaptic cleft. While all TCAs inhibit these transporters, they do so at varying levels (Table 336-51). Based on their chemical structure, TCAs can be categorized into secondary and tertiary amines. Tertiary amines are metabolized via demethylation into their derivatives (Table 336-51). Patients who have recently suffered a myocardial infarction (MI) should avoid tertiary amines. TCAs can reduce heart rate variability, which is already decreased after an MI, thus presenting the potential for cardiac arrhythmias. TCAs should also be avoided in patients with cardiac conduction abnormalities.38-46,52 Patients with a prior baseline cardiac conduction defect, such as a bundle branch block, are at higher risk for further cardiac abnormalities. In those with a preexisting first-degree heart block, TCAs can still be used, but electrocardiogram monitoring is recommended.52,53 TCAs have also been reported to decrease the seizure threshold.38-46 They can be used with caution in patients who have a history of epilepsy or head trauma, or with concomitant medications that lower the seizure threshold.38-46

CP02212035_t3.png

Overdose risk is a concern with TCAs because ingestion of 10 to 20 mg/kg can lead to significant toxicity.54 This is due to their blockage of voltage-gated sodium channels found in the CNS and heart, which contributes to overdose symptoms such as a widened QRS complex and seizures. Symptoms usually develop within 2 hours but may be delayed up to 6 hours.55 Patients with a history of overdose must be carefully assessed before initiating a TCA. Prescribing a limited supply of these medications may be valuable. The use of TCAs has often been limited due to their adverse effects, most of which are associated with their respective affinities for alpha 1, muscarinic 1, and histamine 1 receptors. Inhibition of the alpha 1 receptor is associated with hypotension, muscarinic 1 with anticholinergic adverse effects, and histamine 1 with sedation and weight gain. Tertiary amines have a higher affinity for these receptors compared to secondary amines, leading to a more significant adverse effect profile.36,50 Among TCAs, amitriptyline is the most likely to cause hypotension, whereas desipramine and nortriptyline are least likely. Amitriptyline and clomipramine are most likely to cause anticholinergic adverse effects, whereas desipramine and nortriptyline are the least likely. Amitriptyline, doxepin, and imipramine have the highest propensity for QTc prolongation.36

Beyond treating MDD, TCAs have shown benefits for treating other disease states (Table 438-46,49,56-61).These differing indications may help psychiatrists determine the best TCA to prescribe for a given patient. Amitriptyline is the most studied TCA for MDD; however, nortriptyline is typically preferred due to its favorable tolerability profile.4,62 Nortriptyline also has data supporting its use in ECT to prevent relapse.63 Amitriptyline and nortriptyline have shown benefits in patients with neuro­pathic pain and for migraine prophylaxis.56-60 Although frequently used for MDD, clomipramine is not FDA-approved for this indication, but is for obsessive-compulsive disorder.39 Doxepin is FDA-approved for insomnia at lower doses and for MDD at higher doses.40 Therefore, it may benefit patients with sleep difficulties secondary to depression. Desipramine has been used off-label to treat ADHD in children and has shown some benefits in adults.64-66 Protriptyline, trimipramine, and amoxapine are infrequently used in clinical practice.

CP02212035_t4.png

A unique feature of TCAs is the ability to monitor serum concentrations (Table 336-51).Guidelines recommend therapeutic drug monitoring (TDM) with amitriptyline, clomi­pramine, imipramine, and nortriptyline for routine use. TDM is still recommended for doxepin, desipramine, and trimipramine, but its utility is largely for treatment failure or resistance.37 These plasma levels can be altered based on coadministered medications (Table 538-46) and should be closely monitored. Physicians should obtain a trough level after at least 5 half-lives and before the next dose is due, and use TDM as indicated to optimize dosing.

CP02212035_t5.png

Continue to: CASE CONTINUED

 

 

CASE CONTINUED

Ms. B’s outpatient psychiatrist provides collateral information about her medical history and confirms her long-standing MDD with multiple medication trials, though she has never received an MAOI or TCA. Ms. B is adamant she does not want a medication-free period between treatments and refuses to adjust her diet, despite being educated on the few changes necessary. She has no contraindications for TCAs and may benefit from a TCA for her comorbid migraines. The care team expresses concern for TCA overdose to Ms. B and her family. Ms. B’s sister reassures the team they will have someone monitor and dispense her medications at home. They decide to discontinue her current psychiatric regimen, and Ms. B is started on nortriptyline 50 mg/d at night, with plans to titrate based on tolerability.

Related Resources

Drug Brand Names

Amitriptyline • Elavil
Amphetamine • Adzenys, Dyanavel
Aripiprazole • Abilify
Clomipramine • Anafranil
Desipramine • Norpramin
Dextromethorphan/bupropion • Auvelity
Doxepin • Sinequan, Adapin
Esketamine • Spravato
Fluoxetine • Prozac
Imipramine • Tofranil
Isocarboxazid • Marplan
Methamphetamine • Desoxyn
Mirtazapine • Remeron
Modafinil • Provigil
Nortriptyline • Pamelor
Phenelzine • Nardil
Protriptyline • Vivactil
Selegiline • Emsam
Tranylcypromine • Parnate
Trimipramine • Surmontil
Vortioxetine • Trintellix

[embed:render:related:node:153015]

[embed:render:related:node:246706]

CP02212035_pp.png

Ms. B, age 45, has a history of major depressive disorder (MDD) and migraines. She is admitted after presenting with anhedonia, hopelessness, and hypersomnia. These symptoms have become more severe over the last few weeks. Ms. B describes a past suicide attempt via overdose on doxylamine for which she required treatment in the intensive care unit. The only activity she enjoys is her weekly girls’ night, during which she drinks a few glasses of wine. Ms. B’s current medications are dextromethorphan/bupropion 45/105 mg twice daily and aripiprazole 5 mg/d, which she has taken for 3 months. She states she has “been on every antidepressant there is.”

When clinicians review Ms. B’s medication history, it is clear she has had adequate trials of selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), intranasal esketamine, multiple augmentation strategies, and electroconvulsive therapy (ECT). Ms. B seeks an alternative medication to improve her depressive symptoms.

Treatment-resistant depression (TRD) is commonly defined as depression that has not responded to ≥2 adequate trials of an antidepressant.1 Some guidelines recommend monoamine oxidase inhibitors (MAOIs) and tricyclic antidepressants (TCAs) as second- or even third-line options for MDD,2 while others recommend reserving them for patients with insufficient responses to alternative treatment modalities.3,4 Although MAOIs and TCAs have been available since the 1950s, prescribing these medications has become less prevalent due to safety concerns, the availability of other pharmacologic options, and a lack of clinical training and comfort.5,6 Most research notes that MAOIs are superior for treating atypical depression while TCAs are more effective for melancholic depression.2-4 In a review of 20 studies, Thase et al7 found that 50% of TCA nonresponders benefited from an MAOI. In the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) trial, monotherapy with the MAOI tranylcypromine was associated with a lower remission rate than the TCA nortriptyline; many argue the dose of tranylcypromine was suboptimal, and few participants completed an adequate trial in the last level.8,9 A more recent study by Kim et al10 found MAOIs to be “generally more effective” than TCAs for TRD, particularly in patients with fewer antidepressant trials; however, this was a small retrospective exploratory trial. A network meta-analysis found both classes to be “competitive” with SSRIs based on efficacy and tolerability, which leads to the question of whether these medications should be considered earlier in therapy.11 Considering patient-specific factors and particular medication properties is an effective strategy when prescribing an MAOI or TCA.

Monoamine oxidase inhibitors

Four MAOIs are FDA-approved for treating MDD (Table 15,12-17): phenelzine, isocarboxazid, tranylcypromine, and selegiline. These medications irreversibly inhibit MAO, which exists as isomers A and B. MAO-A primarily metabolizes serotonin and norepinephrine, which is largely responsible for these medications’ antidepressant effects. Both isomers equally metabolize dopamine.5,12,18 It is best to avoid using MAOIs in patients with cerebrovascular disease, hepatic disease, or pheochromocytoma. Patients with active substance use disorders (particularly sympathomimetics and hallucinogens) are at an increased risk for hypertensive crises and serotonin syndrome, respectively. The most common adverse effects are orthostatic hypotension (despite more well-known concerns regarding hypertension), alterations in sleep patterns (insomnia or hypersomnia, depending on the agent), gastrointestinal issues, and anticholinergic adverse effects such as dry mouth and constipation.13,19-21

CP02212035_t1.png

In one review and meta-analysis, phenelzine displayed the highest efficacy across all MAOIs.11 It likely requires high doses to achieve adequate MAO inhibition.11 A metabolite of phenelzine inhibits gamma-aminobutyric acid transaminase and may be helpful for patients with comorbid anxiety disorders or MDD with anxious distress.18,21 Additional considerations include phenelzine’s propensity for orthostasis (with rapid titrations and higher doses), sedation, weight gain, sexual dysfunction, and a rare adverse effect of vitamin B6 deficiency.5,13,14,20-22

Use of isocarboxazid in clinical practice is rare. Its adverse effects are similar to those of phenelzine but isocarboxazid is less studied. Tranylcypromine has a similar chemical structure to amphetamine. It can be stimulating at higher doses, potentially benefitting patients with comorbid attention-deficit/hyperactivity disorder (ADHD) or significant apathy.13,23 Selegiline’s distinct quality is its availability as a transdermal patch, which may be useful for patients who struggle to take oral medications. At low doses (6 mg/24 h), the selegiline transdermal patch allows patients to disregard a dietary tyramine restriction because it avoids first-pass metabolism. It inhibits both MAO isomers in the brain but is only selective for MAO-B once concentrations are distributed to the liver. Higher doses require a tyramine-restricted diet because there is still some MAO-A inhibition in the gut. Selegiline is also stimulating because it is converted to amphetamine and methamphetamine.5,12,13,17,19,24

Despite promising results from the use of MAOIs, physicians and patients may be reluctant to use these medications due to perceived limitations. One prominent barrier is the infamous “cheese reaction.” Tyramine, an amino acid found in certain food and beverages (Table 25,13-18,25-28), is broken down by MAO-A in the gut. When this enzyme is inhibited, higher concentrations of tyramine reach systemic circulation. Tyramine’s release of norepinephrine (which now cannot be broken down) can lead to a hypertensive crisis. Consequently, a tyramine-restricted diet is recommended for patients taking an MAOI. However, the common notion that cheese, wine, and beer must be avoided is false, because most of the dietary restrictions developed following the discovery of MAOIs are antiquated.5,12,25-28 Patients who take an MAOI only need to slightly adjust their diet, as outlined in Table 2.5,13-18,25-28 A reasonable serving size of most foods and beverages containing tyramine is unlikely to elicit this “pressor” response. Of the 4 MAOIs FDA-approved for MDD, tranylcypromine appears to be the most sensitive to tyramine.21 Transient postdose hypertension (regardless of tyramine) may occur after taking an MAOI.29 Encourage patients to monitor their blood pressure.

CP02212035_t2.png

Continue to: Additional hurdles include...

 

 

Additional hurdles include the required washout period from serotonergic medications and interactions with sympathomimetics. MAOIs pose the highest risk of serotonin syndrome; however, this usually occurs if given concomitantly with other serotonergic agents. The standard recommendation is a 14-day washout period from SSRIs (5 weeks for fluoxetine and 3 weeks for vortioxetine), SNRIs, mirtazapine, and other antidepressants. It can be distressing for patients to be without medication during that period. Because some antidepressants have much shorter half-lives, waiting 5 half-lives (typically 5 to 7 days) for the discontinued medication to be excreted is feasible if patients are closely monitored.5,12,13,25,27,30 There are rare instances where a TCA may be combined with an MAOI (typically initiated within 1 to 2 days of each other), but never clomipramine or imipramine due to their potent serotonin reuptake inhibition.31 If switching to an alternative MAOI, waiting 7 to 14 days is recommended to allow adequate time for the inhibited enzyme to regenerate.14-17,32 Taking medications that increase dopamine and norepinephrine (eg, stimulants or oral over-the-counter decongestants) with an MAOI is typically not recommended due to the risk of hypertensive crisis.25,27 In severe TRD or comorbid ADHD, successful simultaneous use of methylphenidate or amphetamine—typically at low doses—with close blood pressure monitoring has been reported.33 There have also been positive cases of the use of modafinil in combination with an MAOI; however, this should be done with caution.34,35 Clinicians must use clinical judgment when considering a combination of medications that pose a higher risk.

Tricyclic antidepressants

TCAs work differently than MAOIs to increase monoamines. They inhibit presynaptic serotonin and norepinephrine transporters in the CNS to increase levels of these chemicals in the synaptic cleft. While all TCAs inhibit these transporters, they do so at varying levels (Table 336-51). Based on their chemical structure, TCAs can be categorized into secondary and tertiary amines. Tertiary amines are metabolized via demethylation into their derivatives (Table 336-51). Patients who have recently suffered a myocardial infarction (MI) should avoid tertiary amines. TCAs can reduce heart rate variability, which is already decreased after an MI, thus presenting the potential for cardiac arrhythmias. TCAs should also be avoided in patients with cardiac conduction abnormalities.38-46,52 Patients with a prior baseline cardiac conduction defect, such as a bundle branch block, are at higher risk for further cardiac abnormalities. In those with a preexisting first-degree heart block, TCAs can still be used, but electrocardiogram monitoring is recommended.52,53 TCAs have also been reported to decrease the seizure threshold.38-46 They can be used with caution in patients who have a history of epilepsy or head trauma, or with concomitant medications that lower the seizure threshold.38-46

CP02212035_t3.png

Overdose risk is a concern with TCAs because ingestion of 10 to 20 mg/kg can lead to significant toxicity.54 This is due to their blockage of voltage-gated sodium channels found in the CNS and heart, which contributes to overdose symptoms such as a widened QRS complex and seizures. Symptoms usually develop within 2 hours but may be delayed up to 6 hours.55 Patients with a history of overdose must be carefully assessed before initiating a TCA. Prescribing a limited supply of these medications may be valuable. The use of TCAs has often been limited due to their adverse effects, most of which are associated with their respective affinities for alpha 1, muscarinic 1, and histamine 1 receptors. Inhibition of the alpha 1 receptor is associated with hypotension, muscarinic 1 with anticholinergic adverse effects, and histamine 1 with sedation and weight gain. Tertiary amines have a higher affinity for these receptors compared to secondary amines, leading to a more significant adverse effect profile.36,50 Among TCAs, amitriptyline is the most likely to cause hypotension, whereas desipramine and nortriptyline are least likely. Amitriptyline and clomipramine are most likely to cause anticholinergic adverse effects, whereas desipramine and nortriptyline are the least likely. Amitriptyline, doxepin, and imipramine have the highest propensity for QTc prolongation.36

Beyond treating MDD, TCAs have shown benefits for treating other disease states (Table 438-46,49,56-61).These differing indications may help psychiatrists determine the best TCA to prescribe for a given patient. Amitriptyline is the most studied TCA for MDD; however, nortriptyline is typically preferred due to its favorable tolerability profile.4,62 Nortriptyline also has data supporting its use in ECT to prevent relapse.63 Amitriptyline and nortriptyline have shown benefits in patients with neuro­pathic pain and for migraine prophylaxis.56-60 Although frequently used for MDD, clomipramine is not FDA-approved for this indication, but is for obsessive-compulsive disorder.39 Doxepin is FDA-approved for insomnia at lower doses and for MDD at higher doses.40 Therefore, it may benefit patients with sleep difficulties secondary to depression. Desipramine has been used off-label to treat ADHD in children and has shown some benefits in adults.64-66 Protriptyline, trimipramine, and amoxapine are infrequently used in clinical practice.

CP02212035_t4.png

A unique feature of TCAs is the ability to monitor serum concentrations (Table 336-51).Guidelines recommend therapeutic drug monitoring (TDM) with amitriptyline, clomi­pramine, imipramine, and nortriptyline for routine use. TDM is still recommended for doxepin, desipramine, and trimipramine, but its utility is largely for treatment failure or resistance.37 These plasma levels can be altered based on coadministered medications (Table 538-46) and should be closely monitored. Physicians should obtain a trough level after at least 5 half-lives and before the next dose is due, and use TDM as indicated to optimize dosing.

CP02212035_t5.png

Continue to: CASE CONTINUED

 

 

CASE CONTINUED

Ms. B’s outpatient psychiatrist provides collateral information about her medical history and confirms her long-standing MDD with multiple medication trials, though she has never received an MAOI or TCA. Ms. B is adamant she does not want a medication-free period between treatments and refuses to adjust her diet, despite being educated on the few changes necessary. She has no contraindications for TCAs and may benefit from a TCA for her comorbid migraines. The care team expresses concern for TCA overdose to Ms. B and her family. Ms. B’s sister reassures the team they will have someone monitor and dispense her medications at home. They decide to discontinue her current psychiatric regimen, and Ms. B is started on nortriptyline 50 mg/d at night, with plans to titrate based on tolerability.

Related Resources

Drug Brand Names

Amitriptyline • Elavil
Amphetamine • Adzenys, Dyanavel
Aripiprazole • Abilify
Clomipramine • Anafranil
Desipramine • Norpramin
Dextromethorphan/bupropion • Auvelity
Doxepin • Sinequan, Adapin
Esketamine • Spravato
Fluoxetine • Prozac
Imipramine • Tofranil
Isocarboxazid • Marplan
Methamphetamine • Desoxyn
Mirtazapine • Remeron
Modafinil • Provigil
Nortriptyline • Pamelor
Phenelzine • Nardil
Protriptyline • Vivactil
Selegiline • Emsam
Tranylcypromine • Parnate
Trimipramine • Surmontil
Vortioxetine • Trintellix

[embed:render:related:node:153015]

[embed:render:related:node:246706]

References

1. Gaynes BN, Lux L, Gartlehner G, et al. Defining treatment-resistant depression. Depress Anxiety. 2020;37(2):134-145. doi:10.1002/da.22968

2. Kennedy SH, Lam RW, McIntyre RS, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 clinical guidelines for the management of adults with major depressive disorder: section 3. Pharmacological treatments. Can J Psychiatry. 2016;61(9):540-560. doi:10.1177/0706743716659417

3. VA/DoD clinical practice guideline for the management of major depressive disorder. Veterans Health Administration and Department of Defense; 2016. https://www.healthquality.va.gov/guidelines/MH/mdd/VADoDMDDCPGFINAL82916.pdf

4. Gelenberg AJ, Freeman MP, Markowitz JC, et al. Practice guideline for the treatment of patients with major depressive disorder. Am J Psychiatry. 2010;167(Suppl 10):9-118.

5. Meyer JM. A concise guide to monoamine oxidase inhibitors. Current Psychiatry. 2017;16(12):14-16,18-23,47,A.

6. Taylor D. Selective serotonin reuptake inhibitors and tricyclic antidepressants in combination. Interactions and therapeutic uses. Br J Psychiatry. 1995;167(5):575-580. doi:10.1192/bjp.167.5.575

7. Thase ME, Trivedi MH, Rush AJ. MAOIs in the contemporary treatment of depression. Neuropsychopharmacology. 1995;12(3):185-219. doi:10.1016/0893-133X(94)00058-8

8. Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006;163(11):1905-1917. doi:10.1176/ajp.2006.163.11.1905

9. McGrath PJ, Stewart JW, Fava M, et al. Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: a STAR*D report. Am J Psychiatry. 2006;163(9):1531-1666. doi:10.1176/ajp.2006.163.9.1531

10. Kim T, Xu C, Amsterdam JD. Relative effectiveness of tricyclic antidepressant versus monoamine oxidase inhibitor monotherapy for treatment-resistant depression. J Affect Disord. 2019;250:199-203. doi:10.1016/j.jad.2019.03.028

11. Suchting R, Tirumalajaru V, Gareeb R, et al. Revisiting monoamine oxidase inhibitors for the treatment of depressive disorders: a systematic review and network meta-analysis. J Affect Disord. 2021;282:1153-1160. doi:10.1016/j.jad.2021.01.021


12. Stahl SM, Felker A. Monoamine oxidase inhibitors: a modern guide to an unrequited class of antidepressants. CNS Spectr. 2008;13(10):855-870. doi:10.1017/s1092852900016965

13. Chamberlain SR, Baldwin DS. Monoamine oxidase inhibitors (MAOIs) in psychiatric practice: how to use them safely and effectively. CNS Drugs. 2021;35(7):703-716. doi:10.1007/s40263-021-00832-x

14. Nardil [package insert]. New York, NY: Parke-Davis; 2009.

15. Marplan [package insert]. Parsippany, NJ: Validus Pharmaceuticals LLC; 2020.

16. Parnate [package insert]. Saint Michael, Barbados: Concordia Pharmaceuticals; 2015.

17. Emsam [package insert]. Morgantown, WV: Mylan Specialty LP; 2014.

18. Shulman KI, Herrmann N, Walker SE. Current place of monoamine oxidase inhibitors in the treatment of depression. CNS Drugs. 2013;27(10):789-797. doi:10.1007/s40263-013-0097-3

19. Sub Laban T, Saadabadi A. Monoamine oxidase inhibitors (MAOI). StatPearls Publishing; 2023. https://www.ncbi.nlm.nih.gov/books/NBK539848/

20. Rabkin JG, Quitkin FM, McGrath P, et al. Adverse reactions to monoamine oxidase inhibitors. Part II. Treatment correlates and clinical management. J Clin Psychopharmacol. 1985;5(1):2-9.

21. Gillman PK. Advances pertaining to the pharmacology and interactions of irreversible nonselective monoamine oxidase inhibitors. J Clin Psychopharmacol. 2011;31(1):66-74. doi:10.1097/JCP.0b013e31820469ea

22. Sidhu G, Marwaha R. Phenelzine. StatPearls Publishing; 2023. https://www.ncbi.nlm.nih.gov/books/NBK554508/

23. Frieling H, Bleich S. Tranylcypromine: new perspectives on an “old” drug. Eur Arch Psychiatry Clin Neurosci. 2006;256(5):268-273. doi:10.1007/s00406-006-0660-8

24. Goodnick PJ. Seligiline transdermal system in depression. Expert Opin Pharmacother. 2007;8(1):59-64. doi:10.1517/14656566.8.1.59

25. Edinoff AN, Swinford CR, Odisho AS, et al. Clinically relevant drug interactions with monoamine oxidase inhibitors. Health Psychol Res. 2022;10(4):39576. doi:10.52965/001c.39576

26. Gillman PK. A reassessment of the safety profile of monoamine oxidase inhibitors: elucidating tired old tyramine myths. J Neural Transm (Vienna). 2018;125(11):1707-1717. doi:10.1007/s00702-018-1932-y

27. Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03

28. McCabe-Sellers BJ, Staggs CG, Bogle ML. Tyramine in foods and monoamine oxidase inhibitor drugs: a crossroad where medicine, nutrition, pharmacy, and food industry converge. J Food Composit Anal. 2006;19:S58-S65. doi:10.1016/j.jfca.2005.12.008

29. Keck PE Jr, Vuckovic A, Pope HG Jr, et al. Acute cardiovascular response to monoamine oxidase inhibitors: a prospective assessment. J Clin Psychopharmacol. 1989;9(3):203-206.

30. Bodkin JA, Dunlop BW. Moving on with monoamine oxidase inhibitors. Focus (Am Psychiatr Publ). 2021;19(1):50-52. doi:10.1176/appi.focus.20200046

31. Amsterdam JD, Kim TT. Relative effectiveness of monoamine oxidase inhibitor and tricyclic antidepressant combination therapy for treatment-resistant depression. J Clin Psychopharmacol. 2019;39(6):649-652. doi:10.1097/JCP.0000000000001130

32. Keks N, Hope J, Keogh S. Switching and stopping antidepressants. Aust Prescr. 2016;39(3):76-83. doi:10.18773/austprescr.2016.039

33. Israel JA. Combining stimulants and monoamine oxidase inhibitors: a reexamination of the literature and a report of a new treatment combination. Prim Care Companion CNS Disord. 2015;17(6):10.4088/PCC.15br01836. doi:10.4088/PCC.15br01836

34. Clemons WE, Makela E, Young J. Concomitant use of modafinil and tranylcypromine in a patient with narcolepsy: a case report. Sleep Med. 2004;5(5):509-511. doi:10.1016/j.sleep.2004.06.006

35. Ashton AK. Modafinil augmentation of phenelzine for residual fatigue in dysthymia. Am J Psychiatry. 2004;161(9):1716-1717. doi:10.1176/appi.ajp.161.9.1716-a

36. O’Donnell JM, Bies RR, Shelton RC. Drug therapy of depression and anxiety disorders. In: Brunton LL, Hilal-Dandan R, Knollmann BC, eds. Goodman & Gilman’s: The Pharmacological Basis of Therapeutics. 13th ed. McGraw Hill; 2017. Accessed June 4, 2023. https://accessanesthesiology.mhmedical.com/content.aspx?bookid=2189&sectionid=169518711

37. Hiemke C, Bergemann N, Clement HW, et al. Consensus guidelines for therapeutic drug monitoring in neuropsychopharmacology: update 2017. Pharmacopsychiatry. 2018;51(1-02):9-62. doi:10.1055/s-0043-116492

38. Amitriptyline hydrochloride [package insert]. East Brunswick, NJ: Unichem Pharmaceuticals (USA); 2021.

39. Clomipramine hydrochloride [package insert]. East Windsor, NJ: Aurobindo Pharma Limited; 2023.

40. Doxepin hydrochloride capsules, USP [package insert]. Bedminster, NJ: Alembic Pharmaceuticals Inc; 2021. 

41. Imipramine hydrochloride tablet [package insert]. Fairfield, NJ: Leading Pharma LLC USA; 2022.  

42. Trimipramine maleate [package insert]. Northvale, NJ: Elite Laboratories Inc; 2021. 

43. Amoxapine [package insert]. Parsippany, NJ: Actavis Pharma Inc; 2015.

44. Desipramine hydrochloride tablets [package insert]. Bedminster, NJ: Alembic Pharmaceuticals Inc; 2023.

45. Nortriptyline hydrochloride capsules, USP [package insert]. Parsippany, NJ: Teva Pharmaceuticals Inc; 2021.

46. Protriptyline hydrochloride [package insert]. Bensalem, PA: Sigmapharm Laboratories, LLC; 2023.

47. Calvo B, García MJ, Pedraz JL, et al. Pharmacokinetics of amoxapine and its active metabolites. Int J Clin Pharmacol Ther Toxicol. 1985;23(4):180-185.

48. Ziegler VE, Biggs JT, Wylie LT, et al. Protriptyline kinetics. Clin Pharmacol Ther. 1978;23(5):580-584. doi:10.1002/cpt1978235580

49. Cleare A, Pariante CM, Young AH, et al. Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol. 2015;29(5):459-525. doi:10.1177/0269881115581093

50. Richelson E. Synaptic effects of antidepressants. J Clin Psychopharmacol. 1996;16(3 Suppl 2):1S-9S. doi:10.1097/00004714-199606002-00001

51. Vos CF, Aarnoutse RE, Op de Coul MJM, et al. Tricyclic antidepressants for major depressive disorder: a comprehensive evaluation of current practice in the Netherlands. BMC Psychiatry. 2021;21(1):481. doi:10.1186/s12888-021-03490-x

52. Alvarez W Jr, Pickworth KK. Safety of antidepressant drugs in the patient with cardiac disease: a review of the literature. Pharmacotherapy. 2003;23(6):754-771. doi:10.1592/phco.23.6.754.32185

53. Dietch JT, Fine M. The effect of nortriptyline in elderly patients with cardiac conduction disease. J Clin Psychiatry. 1990;51(2):65-67.

54. Valento M, Liebelt EL. Cyclic antidepressants. In: Nelson LS, Howland M, Lewin NA, et al, eds. Goldfrank’s Toxicologic Emergencies. 9th ed. McGraw Hill; 2011. Accessed June 10, 2023. https://accesspharmacy.mhmedical.com/content.aspx?bookid=2569&sectionid=210274664

55. Woolf AD, Erdman AR, Nelson LS, et al. Tricyclic antidepressant poisoning: an evidence-based consensus guideline for out-of-hospital management. Clin Toxicol (Phila). 2007;45(3):203-233. doi:10.1080/15563650701226192

56. Baldwin DS, Anderson IM, Nutt DJ, et al. Evidence-based pharmacological treatment of anxiety disorders, post-traumatic stress disorder and obsessive-compulsive disorder: a revision of the 2005 guidelines from the British Association for Psychopharmacology. J Psychopharmacol. 2014;28(5):403-439. doi:10.1177/0269881114525674

57. Moulin DE, Clark AJ, Gilron I, et al. Pharmacological management of chronic neuropathic pain - consensus statement and guidelines from the Canadian Pain Society. Pain Res Manag. 2007;12(1):13-21. doi:10.1155/2007/730785

58. Finnerup NB, Attal N, Haroutounian S, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015;14(2):162-173. doi:10.1016/S1474-4422(14)70251-0

59. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev. 2007;(4):CD005454. doi:10.1002/14651858.CD005454.pub2

60. Burch R. Antidepressants for preventive treatment of migraine. Curr Treat Options Neurol. 2019;21(4):18. doi:10.1007/s11940-019-0557-2

61. Ailani J, Burch RC, Robbins MS; Board of Directors of the American Headache Society. The American Headache Society consensus statement: update on integrating new migraine treatments into clinical practice. Headache. 2021;61(7):1021-1039. doi:10.1111/head.14153

62. Bauer M, Pfennig A, Severus E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for biological treatment of unipolar depressive disorders, part 1: update 2013 on the acute and continuation treatment of unipolar depressive disorders. World J Biol Psychiatry. 2013;14(5):334-385. doi:10.3109/15622975.2013.804195

63. Jelovac A, Kolshus E, McLoughlin DM. Relapse following successful electroconvulsive therapy for major depression: a meta-analysis. Neuropsychopharmacology. 2013;38(12):2467-2474. doi:10.1038/npp.2013.149

64. Spencer T, Biederman J, Coffey B, et al. A double-blind comparison of desipramine and placebo in children and adolescents with chronic tic disorder and comorbid attention-deficit/hyperactivity disorder. Arch Gen Psychiatry. 2002;59(7):649-656. doi:10.1001/archpsyc.59.7.649

65. Spencer T, Biederman J, Wilens T, et al. Pharmacotherapy of attention-deficit hyperactivity disorder across the life cycle. J Am Acad Child Adolesc Psychiatry. 1996;35(4):409-432. doi:10.1097/00004583-199604000-00008

66. Wilens TE, Biederman J, Prince J, et al. Six-week, double-blind, placebo-controlled study of desipramine for adult attention deficit hyperactivity disorder. Am J Psychiatry. 1996;153(9):1147-1153. doi:10.1176/ajp.153.9.1147

References

1. Gaynes BN, Lux L, Gartlehner G, et al. Defining treatment-resistant depression. Depress Anxiety. 2020;37(2):134-145. doi:10.1002/da.22968

2. Kennedy SH, Lam RW, McIntyre RS, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 clinical guidelines for the management of adults with major depressive disorder: section 3. Pharmacological treatments. Can J Psychiatry. 2016;61(9):540-560. doi:10.1177/0706743716659417

3. VA/DoD clinical practice guideline for the management of major depressive disorder. Veterans Health Administration and Department of Defense; 2016. https://www.healthquality.va.gov/guidelines/MH/mdd/VADoDMDDCPGFINAL82916.pdf

4. Gelenberg AJ, Freeman MP, Markowitz JC, et al. Practice guideline for the treatment of patients with major depressive disorder. Am J Psychiatry. 2010;167(Suppl 10):9-118.

5. Meyer JM. A concise guide to monoamine oxidase inhibitors. Current Psychiatry. 2017;16(12):14-16,18-23,47,A.

6. Taylor D. Selective serotonin reuptake inhibitors and tricyclic antidepressants in combination. Interactions and therapeutic uses. Br J Psychiatry. 1995;167(5):575-580. doi:10.1192/bjp.167.5.575

7. Thase ME, Trivedi MH, Rush AJ. MAOIs in the contemporary treatment of depression. Neuropsychopharmacology. 1995;12(3):185-219. doi:10.1016/0893-133X(94)00058-8

8. Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006;163(11):1905-1917. doi:10.1176/ajp.2006.163.11.1905

9. McGrath PJ, Stewart JW, Fava M, et al. Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: a STAR*D report. Am J Psychiatry. 2006;163(9):1531-1666. doi:10.1176/ajp.2006.163.9.1531

10. Kim T, Xu C, Amsterdam JD. Relative effectiveness of tricyclic antidepressant versus monoamine oxidase inhibitor monotherapy for treatment-resistant depression. J Affect Disord. 2019;250:199-203. doi:10.1016/j.jad.2019.03.028

11. Suchting R, Tirumalajaru V, Gareeb R, et al. Revisiting monoamine oxidase inhibitors for the treatment of depressive disorders: a systematic review and network meta-analysis. J Affect Disord. 2021;282:1153-1160. doi:10.1016/j.jad.2021.01.021


12. Stahl SM, Felker A. Monoamine oxidase inhibitors: a modern guide to an unrequited class of antidepressants. CNS Spectr. 2008;13(10):855-870. doi:10.1017/s1092852900016965

13. Chamberlain SR, Baldwin DS. Monoamine oxidase inhibitors (MAOIs) in psychiatric practice: how to use them safely and effectively. CNS Drugs. 2021;35(7):703-716. doi:10.1007/s40263-021-00832-x

14. Nardil [package insert]. New York, NY: Parke-Davis; 2009.

15. Marplan [package insert]. Parsippany, NJ: Validus Pharmaceuticals LLC; 2020.

16. Parnate [package insert]. Saint Michael, Barbados: Concordia Pharmaceuticals; 2015.

17. Emsam [package insert]. Morgantown, WV: Mylan Specialty LP; 2014.

18. Shulman KI, Herrmann N, Walker SE. Current place of monoamine oxidase inhibitors in the treatment of depression. CNS Drugs. 2013;27(10):789-797. doi:10.1007/s40263-013-0097-3

19. Sub Laban T, Saadabadi A. Monoamine oxidase inhibitors (MAOI). StatPearls Publishing; 2023. https://www.ncbi.nlm.nih.gov/books/NBK539848/

20. Rabkin JG, Quitkin FM, McGrath P, et al. Adverse reactions to monoamine oxidase inhibitors. Part II. Treatment correlates and clinical management. J Clin Psychopharmacol. 1985;5(1):2-9.

21. Gillman PK. Advances pertaining to the pharmacology and interactions of irreversible nonselective monoamine oxidase inhibitors. J Clin Psychopharmacol. 2011;31(1):66-74. doi:10.1097/JCP.0b013e31820469ea

22. Sidhu G, Marwaha R. Phenelzine. StatPearls Publishing; 2023. https://www.ncbi.nlm.nih.gov/books/NBK554508/

23. Frieling H, Bleich S. Tranylcypromine: new perspectives on an “old” drug. Eur Arch Psychiatry Clin Neurosci. 2006;256(5):268-273. doi:10.1007/s00406-006-0660-8

24. Goodnick PJ. Seligiline transdermal system in depression. Expert Opin Pharmacother. 2007;8(1):59-64. doi:10.1517/14656566.8.1.59

25. Edinoff AN, Swinford CR, Odisho AS, et al. Clinically relevant drug interactions with monoamine oxidase inhibitors. Health Psychol Res. 2022;10(4):39576. doi:10.52965/001c.39576

26. Gillman PK. A reassessment of the safety profile of monoamine oxidase inhibitors: elucidating tired old tyramine myths. J Neural Transm (Vienna). 2018;125(11):1707-1717. doi:10.1007/s00702-018-1932-y

27. Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03

28. McCabe-Sellers BJ, Staggs CG, Bogle ML. Tyramine in foods and monoamine oxidase inhibitor drugs: a crossroad where medicine, nutrition, pharmacy, and food industry converge. J Food Composit Anal. 2006;19:S58-S65. doi:10.1016/j.jfca.2005.12.008

29. Keck PE Jr, Vuckovic A, Pope HG Jr, et al. Acute cardiovascular response to monoamine oxidase inhibitors: a prospective assessment. J Clin Psychopharmacol. 1989;9(3):203-206.

30. Bodkin JA, Dunlop BW. Moving on with monoamine oxidase inhibitors. Focus (Am Psychiatr Publ). 2021;19(1):50-52. doi:10.1176/appi.focus.20200046

31. Amsterdam JD, Kim TT. Relative effectiveness of monoamine oxidase inhibitor and tricyclic antidepressant combination therapy for treatment-resistant depression. J Clin Psychopharmacol. 2019;39(6):649-652. doi:10.1097/JCP.0000000000001130

32. Keks N, Hope J, Keogh S. Switching and stopping antidepressants. Aust Prescr. 2016;39(3):76-83. doi:10.18773/austprescr.2016.039

33. Israel JA. Combining stimulants and monoamine oxidase inhibitors: a reexamination of the literature and a report of a new treatment combination. Prim Care Companion CNS Disord. 2015;17(6):10.4088/PCC.15br01836. doi:10.4088/PCC.15br01836

34. Clemons WE, Makela E, Young J. Concomitant use of modafinil and tranylcypromine in a patient with narcolepsy: a case report. Sleep Med. 2004;5(5):509-511. doi:10.1016/j.sleep.2004.06.006

35. Ashton AK. Modafinil augmentation of phenelzine for residual fatigue in dysthymia. Am J Psychiatry. 2004;161(9):1716-1717. doi:10.1176/appi.ajp.161.9.1716-a

36. O’Donnell JM, Bies RR, Shelton RC. Drug therapy of depression and anxiety disorders. In: Brunton LL, Hilal-Dandan R, Knollmann BC, eds. Goodman & Gilman’s: The Pharmacological Basis of Therapeutics. 13th ed. McGraw Hill; 2017. Accessed June 4, 2023. https://accessanesthesiology.mhmedical.com/content.aspx?bookid=2189&sectionid=169518711

37. Hiemke C, Bergemann N, Clement HW, et al. Consensus guidelines for therapeutic drug monitoring in neuropsychopharmacology: update 2017. Pharmacopsychiatry. 2018;51(1-02):9-62. doi:10.1055/s-0043-116492

38. Amitriptyline hydrochloride [package insert]. East Brunswick, NJ: Unichem Pharmaceuticals (USA); 2021.

39. Clomipramine hydrochloride [package insert]. East Windsor, NJ: Aurobindo Pharma Limited; 2023.

40. Doxepin hydrochloride capsules, USP [package insert]. Bedminster, NJ: Alembic Pharmaceuticals Inc; 2021. 

41. Imipramine hydrochloride tablet [package insert]. Fairfield, NJ: Leading Pharma LLC USA; 2022.  

42. Trimipramine maleate [package insert]. Northvale, NJ: Elite Laboratories Inc; 2021. 

43. Amoxapine [package insert]. Parsippany, NJ: Actavis Pharma Inc; 2015.

44. Desipramine hydrochloride tablets [package insert]. Bedminster, NJ: Alembic Pharmaceuticals Inc; 2023.

45. Nortriptyline hydrochloride capsules, USP [package insert]. Parsippany, NJ: Teva Pharmaceuticals Inc; 2021.

46. Protriptyline hydrochloride [package insert]. Bensalem, PA: Sigmapharm Laboratories, LLC; 2023.

47. Calvo B, García MJ, Pedraz JL, et al. Pharmacokinetics of amoxapine and its active metabolites. Int J Clin Pharmacol Ther Toxicol. 1985;23(4):180-185.

48. Ziegler VE, Biggs JT, Wylie LT, et al. Protriptyline kinetics. Clin Pharmacol Ther. 1978;23(5):580-584. doi:10.1002/cpt1978235580

49. Cleare A, Pariante CM, Young AH, et al. Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol. 2015;29(5):459-525. doi:10.1177/0269881115581093

50. Richelson E. Synaptic effects of antidepressants. J Clin Psychopharmacol. 1996;16(3 Suppl 2):1S-9S. doi:10.1097/00004714-199606002-00001

51. Vos CF, Aarnoutse RE, Op de Coul MJM, et al. Tricyclic antidepressants for major depressive disorder: a comprehensive evaluation of current practice in the Netherlands. BMC Psychiatry. 2021;21(1):481. doi:10.1186/s12888-021-03490-x

52. Alvarez W Jr, Pickworth KK. Safety of antidepressant drugs in the patient with cardiac disease: a review of the literature. Pharmacotherapy. 2003;23(6):754-771. doi:10.1592/phco.23.6.754.32185

53. Dietch JT, Fine M. The effect of nortriptyline in elderly patients with cardiac conduction disease. J Clin Psychiatry. 1990;51(2):65-67.

54. Valento M, Liebelt EL. Cyclic antidepressants. In: Nelson LS, Howland M, Lewin NA, et al, eds. Goldfrank’s Toxicologic Emergencies. 9th ed. McGraw Hill; 2011. Accessed June 10, 2023. https://accesspharmacy.mhmedical.com/content.aspx?bookid=2569&sectionid=210274664

55. Woolf AD, Erdman AR, Nelson LS, et al. Tricyclic antidepressant poisoning: an evidence-based consensus guideline for out-of-hospital management. Clin Toxicol (Phila). 2007;45(3):203-233. doi:10.1080/15563650701226192

56. Baldwin DS, Anderson IM, Nutt DJ, et al. Evidence-based pharmacological treatment of anxiety disorders, post-traumatic stress disorder and obsessive-compulsive disorder: a revision of the 2005 guidelines from the British Association for Psychopharmacology. J Psychopharmacol. 2014;28(5):403-439. doi:10.1177/0269881114525674

57. Moulin DE, Clark AJ, Gilron I, et al. Pharmacological management of chronic neuropathic pain - consensus statement and guidelines from the Canadian Pain Society. Pain Res Manag. 2007;12(1):13-21. doi:10.1155/2007/730785

58. Finnerup NB, Attal N, Haroutounian S, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015;14(2):162-173. doi:10.1016/S1474-4422(14)70251-0

59. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev. 2007;(4):CD005454. doi:10.1002/14651858.CD005454.pub2

60. Burch R. Antidepressants for preventive treatment of migraine. Curr Treat Options Neurol. 2019;21(4):18. doi:10.1007/s11940-019-0557-2

61. Ailani J, Burch RC, Robbins MS; Board of Directors of the American Headache Society. The American Headache Society consensus statement: update on integrating new migraine treatments into clinical practice. Headache. 2021;61(7):1021-1039. doi:10.1111/head.14153

62. Bauer M, Pfennig A, Severus E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for biological treatment of unipolar depressive disorders, part 1: update 2013 on the acute and continuation treatment of unipolar depressive disorders. World J Biol Psychiatry. 2013;14(5):334-385. doi:10.3109/15622975.2013.804195

63. Jelovac A, Kolshus E, McLoughlin DM. Relapse following successful electroconvulsive therapy for major depression: a meta-analysis. Neuropsychopharmacology. 2013;38(12):2467-2474. doi:10.1038/npp.2013.149

64. Spencer T, Biederman J, Coffey B, et al. A double-blind comparison of desipramine and placebo in children and adolescents with chronic tic disorder and comorbid attention-deficit/hyperactivity disorder. Arch Gen Psychiatry. 2002;59(7):649-656. doi:10.1001/archpsyc.59.7.649

65. Spencer T, Biederman J, Wilens T, et al. Pharmacotherapy of attention-deficit hyperactivity disorder across the life cycle. J Am Acad Child Adolesc Psychiatry. 1996;35(4):409-432. doi:10.1097/00004583-199604000-00008

66. Wilens TE, Biederman J, Prince J, et al. Six-week, double-blind, placebo-controlled study of desipramine for adult attention deficit hyperactivity disorder. Am J Psychiatry. 1996;153(9):1147-1153. doi:10.1176/ajp.153.9.1147

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
35-42
Page Number
35-42
Publications
Publications
Topics
Article Type
Display Headline
Monoamine oxidase inhibitors and tricyclic antidepressants for MDD
Display Headline
Monoamine oxidase inhibitors and tricyclic antidepressants for MDD
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1223 Savvy Costello</fileName> <TBEID>0C02E893.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02E893</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Monoamine oxidase inhibitors and</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231129T083224</firstPublished> <LastPublished>20231129T083225</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231129T083224</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Christie E. Costello, PharmD, BCPS, BCPP</byline> <bylineText/> <bylineFull>Christie E. Costello, PharmD, BCPS, BCPP</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>35-42</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Ms. B, age 45, has a history of major depressive disorder (MDD) and migraines. She is admitted after presenting with anhedonia, hopelessness, and hypersomnia. T</metaDescription> <articlePDF>299297</articlePDF> <teaserImage/> <title>Monoamine oxidase inhibitors and tricyclic antidepressants for MDD</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords> <keyword>depression</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">36771</term> </sections> <topics> <term canonical="true">202</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002645.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Monoamine oxidase inhibitors and tricyclic antidepressants for MDD</title> <deck/> </itemMeta> <itemContent> <p> <strong>Christie E. Costello, PharmD, BCPS, BCPP, and Bridgette M. Gleisner, PharmD, BCPP</strong> </p> <p>Ms. B, age 45, has a history of major depressive disorder (MDD) and migraines. She is admitted after presenting with anhedonia, hopelessness, and hypersomnia. These symptoms have become more severe over the last few weeks. Ms. B describes a past suicide attempt via overdose on doxylamine for which she required treatment in the intensive care unit. The only activity she enjoys is her weekly girls’ night, during which she drinks a few glasses of wine. Ms. B’s current medications are dextromethorphan/bupropion 45/105 mg twice daily and aripiprazole 5 mg/d, which she has taken for 3 months. She states she has “been on every antidepressant there is.”</p> <p>When clinicians review Ms. B’s medication history, it is clear she has had adequate trials of selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), intranasal esketamine, multiple augmentation strategies, and electroconvulsive therapy (ECT). Ms. B seeks an alternative medication to improve her depressive symptoms.</p> <p>Treatment-resistant depression (TRD) is commonly defined as depression that has not responded to ≥2 adequate trials of an antidepressant.<sup>1</sup> Some guidelines recommend monoamine oxidase inhibitors (MAOIs) and tricyclic antidepressants (TCAs) as second- or even third-line options for MDD,<sup>2 </sup>while others recommend reserving them for patients with insufficient responses to alternative treatment modalities.<sup>3,4</sup> Although MAOIs and TCAs have been available since the 1950s, prescribing these medications has become less prevalent due to safety concerns, the availability of other pharmacologic options, and a lack of clinical training and comfort.<sup>5,6</sup> Most research notes that MAOIs are superior for treating atypical depression while TCAs are more effective for melancholic depression.<sup>2-4</sup> In a review of 20 studies, Thase et al<sup>7</sup> found that 50% of TCA nonresponders benefited from an MAOI. In the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) trial, monotherapy with the MAOI tranylcypromine was associated with a lower remission rate than the TCA nortriptyline; many argue the dose of tranylcypromine was suboptimal, and few participants completed an adequate trial in the last level.<sup>8,9</sup> A more recent study by Kim et al<sup>10</sup> found MAOIs to be “generally more effective” than TCAs for TRD, particularly in patients with fewer antidepressant trials; however, this was a small retrospective exploratory trial. A network meta-analysis found both classes to be “competitive” with SSRIs based on efficacy and tolerability, which leads to the question of whether these medications should be considered earlier in therapy.<sup>11</sup> Considering patient-specific factors and particular medication properties is an effective strategy when prescribing an MAOI or TCA.</p> <h3>Monoamine oxidase inhibitors</h3> <p>Four MAOIs are FDA-approved for treating MDD (<span class="bitalic">Table 1</span><sup>5,12-17</sup>): phenelzine, isocarboxazid, tranylcypromine, and selegiline. These medications irreversibly inhibit MAO, which exists as isomers A and B. MAO-A primarily metabolizes serotonin and norepinephrine, which is largely responsible for these medications’ antidepressant effects. Both isomers equally metabolize dopamine.<sup>5,12,18 </sup>It is best to avoid using MAOIs in patients with cerebrovascular disease, hepatic disease, or pheochromocytoma. Patients with active substance use disorders (particularly sympathomimetics and hallucinogens) are at an increased risk for hypertensive crises and serotonin syndrome, respectively. The most common adverse effects are orthostatic hypotension (despite more well-known concerns regarding hypertension), alterations in sleep patterns (insomnia or hypersomnia, depending on the agent), gastrointestinal issues, and anticholinergic adverse effects such as dry mouth and constipation.<sup>13,19-21</sup></p> <p>In one review and meta-analysis, phenelzine displayed the highest efficacy across all MAOIs.<sup>11</sup> It likely requires high doses to achieve adequate MAO inhibition.<sup>11</sup> A metabolite of phenelzine inhibits gamma-aminobutyric acid transaminase and may be helpful for patients with comorbid anxiety disorders or MDD with anxious distress.<sup>18,21</sup> Additional considerations include phenelzine’s propensity for orthostasis (with rapid titrations and higher doses), sedation, weight gain, sexual dysfunction, and a rare adverse effect of vitamin B6 deficiency.<sup>5,13,14,20-22<br/><br/></sup>Use of isocarboxazid in clinical practice is rare. Its adverse effects are similar to those of phenelzine but isocarboxazid is less studied. Tranylcypromine has a similar chemical structure to amphetamine. It can be stimulating at higher doses, potentially benefitting patients with comorbid attention-deficit/hyperactivity disorder (ADHD) or significant apathy.<sup>13,23</sup> Selegiline’s distinct quality is its availability as a transdermal patch, which may be useful for patients who struggle to take oral medications. At low doses (6 mg/24 h), the selegiline transdermal patch allows patients to disregard a dietary tyramine restriction because it avoids first-pass metabolism. It inhibits both MAO isomers in the brain but is only selective for MAO-B once concentrations are distributed to the liver. Higher doses require a tyramine-restricted diet because there is still some MAO-A inhibition in the gut. Selegiline is also stimulating because it is converted to amphetamine and methamphetamine.<sup>5,12,13,17,19,24<br/><br/></sup>Despite promising results from the use of MAOIs, physicians and patients may be reluctant to use these medications due to perceived limitations. One prominent barrier is the infamous “cheese reaction.” Tyramine, an amino acid found in certain food and beverages (<span class="bitalic">Table 2</span>,<sup>5,13-18,25-28</sup> <span class="bitalic">page 36</span>), is broken down by MAO-A in the gut. When this enzyme is inhibited, higher concentrations of tyramine reach systemic circulation. Tyramine’s release of norepinephrine (which now cannot be broken down) can lead to a hypertensive crisis. Consequently, a tyramine-restricted diet is recommended for patients taking an MAOI. However, the common notion that cheese, wine, and beer must be avoided is false, because most of the dietary restrictions developed following the discovery of MAOIs are antiquated.<sup>5,12,25-28 </sup>Patients who take an MAOI only need to slightly adjust their diet, as outlined in <span class="bitalic">Table 2</span>.<sup>5,13-18,25-28</sup> A reasonable serving size of most foods and beverages containing tyramine is unlikely to elicit this “pressor” response. Of the 4 MAOIs FDA-approved for MDD, tranylcypromine appears to be the most sensitive to tyramine.<sup>21</sup> Transient postdose hypertension (regardless of tyramine) may occur after taking an MAOI.<sup>29</sup> Encourage patients to monitor their blood pressure.<br/><br/>Additional hurdles include the required washout period from serotonergic medications and interactions with sympathomimetics. MAOIs pose the highest risk of serotonin syndrome; however, this usually occurs if given concomitantly with other serotonergic agents. The standard recommendation is a 14-day washout period from SSRIs (5 weeks for fluoxetine and 3 weeks for vortioxetine), SNRIs, mirtazapine, and other antidepressants. It can be distressing for patients to be without medication during that period. Because some antidepressants have much shorter half-lives, waiting 5 half-lives (typically 5 to 7 days) for the discontinued medication to be excreted is feasible if patients are closely monitored.<sup>5,12,13,25,27,30</sup> There are rare instances where a TCA may be combined with an MAOI (typically initiated within 1 to 2 days of each other), but never clomipramine or imipramine due to their potent serotonin reuptake inhibition.<sup>31</sup> If switching to an alternative MAOI, waiting 7 to 14 days is recommended to allow adequate time for the inhibited enzyme to regenerate.<sup>14-17,32 </sup>Taking medications that increase dopamine and norepinephrine (eg, stimulants or oral over-the-counter decongestants) with an MAOI is typically not recommended due to the risk of hypertensive crisis.<sup>25,27</sup> In severe TRD or comorbid ADHD, successful simultaneous use of methylphenidate or amphetamine—typically at low doses—with close blood pressure monitoring has been reported.<sup>33</sup> There have also been positive cases of the use of modafinil in combination with an MAOI; however, this should be done with caution.<sup>34,35</sup> Clinicians must use clinical judgment when considering a combination of medications that pose a higher risk.</p> <h3>Tricyclic antidepressants</h3> <p>TCAs work differently than MAOIs to increase monoamines. They inhibit presynaptic serotonin and norepinephrine transporters in the CNS to increase levels of these chemicals in the synaptic cleft. While all TCAs inhibit these transporters, they do so at varying levels (<span class="bitalic">Table 3</span>,<sup>36-51</sup> <span class="bitalic">page 38</span>). Based on their chemical structure, TCAs can be categorized into secondary and tertiary amines. Tertiary amines are metabolized via demethylation into their derivatives (<span class="bitalic">Table 3</span><sup>36-51</sup>). Patients who have recently suffered a myocardial infarction (MI) should avoid tertiary amines. TCAs can reduce heart rate variability, which is already decreased after an MI, thus presenting the potential for cardiac arrhythmias. TCAs should also be avoided in patients with cardiac conduction abnormalities.<sup>38-46,52</sup> Patients with a prior baseline cardiac conduction defect, such as a bundle branch block, are at higher risk for further cardiac abnormalities. In those with a preexisting first-degree heart block, TCAs can still be used, but electrocardiogram monitoring is recommended.<sup>52,53 </sup>TCAs have also been reported to decrease the seizure threshold.<sup>38-46</sup> They can be used with caution in patients who have a history of epilepsy or head trauma, or with concomitant medications that lower the seizure threshold.<sup>38-46</sup></p> <p>Overdose risk is a concern with TCAs because ingestion of 10 to 20 mg/kg can lead to significant toxicity.<sup>54</sup> This is due to their blockage of voltage-gated sodium channels found in the CNS and heart, which contributes to overdose symptoms such as a widened QRS complex and seizures. Symptoms usually develop within 2 hours but may be delayed up to 6 hours.<sup>55</sup> Patients with a history of overdose must be carefully assessed before initiating a TCA. Prescribing a limited supply of these medications may be valuable. The use of TCAs has often been limited due to their adverse effects, most of which are associated with their respective affinities for alpha 1, muscarinic 1, and histamine 1 receptors. Inhibition of the alpha 1 receptor is associated with hypotension, muscarinic 1 with anticholinergic adverse effects, and histamine 1 with sedation and weight gain. Tertiary amines have a higher affinity for these receptors compared to secondary amines, leading to a more significant adverse effect profile.<sup>36,50</sup> Among TCAs, amitriptyline is the most likely to cause hypotension, whereas desipramine and nortriptyline are least likely. Amitriptyline and clomipramine are most likely to cause anticholinergic adverse effects, whereas desipramine and nortriptyline are the least likely. Amitriptyline, doxepin, and imipramine have the highest propensity for QTc prolongation.<sup>36</sup>Beyond treating MDD, TCAs have shown benefits for treating other disease states (<span class="bitalic">Table 4</span>,<sup>38-46,49,56-61</sup> <span class="bitalic">page 39</span>).<sup> </sup>These differing indications may help psychiatrists determine the best TCA to prescribe for a given patient. Amitriptyline is the most studied TCA for MDD; however, nortriptyline is typically preferred due to its favorable tolerability profile.<sup>4,62</sup> Nortriptyline also has data supporting its use in ECT to prevent relapse.<sup>63 </sup>Amitriptyline and nortriptyline have shown benefits in patients with neuro­pathic pain and for migraine prophylaxis.<sup>56-60</sup> Although frequently used for MDD, clomipramine is not FDA-approved for this indication, but is for obsessive-compulsive disorder.<sup>39</sup> Doxepin is FDA-approved for insomnia at lower doses and for MDD at higher doses.<sup>40</sup> Therefore, it may benefit patients with sleep difficulties secondary to depression. Desipramine has been used off-label to treat ADHD in children and has shown some benefits in adults.<sup>64-66</sup> Protriptyline, trimipramine, and amoxapine are infrequently used in clinical practice.<br/><br/>A unique feature of TCAs is the ability to monitor serum concentrations (<span class="bitalic">Table 3</span><sup>36-51</sup>).Guidelines recommend therapeutic drug monitoring (TDM) with amitriptyline, clomi­pramine, imipramine, and nortriptyline for routine use. TDM is still recommended for doxepin, desipramine, and trimipramine, but its utility is largely for treatment failure or resistance.<sup>37</sup> These plasma levels can be altered based on coadministered medications (<b><i>Table 5</i></b>,<sup>38-46</sup> <b><i>page 39</i></b>) and should be closely monitored. Physicians should obtain a trough level after at least 5 half-lives and before the next dose is due, and use TDM as indicated to optimize dosing.</p> <h3> <span class="dingbat"> </span> <span class="box">CASE CONTINUED</span> <span class="dingbat"> </span> </h3> <p>Ms. B’s outpatient psychiatrist provides collateral information about her medical history and confirms her long-standing MDD with multiple medication trials, though she has never received an MAOI or TCA. Ms. B is adamant she does not want a medication-free period between treatments and refuses to adjust her diet, despite being educated on the few changes necessary. She has no contraindications for TCAs and may benefit from a TCA for her comorbid migraines. The care team expresses concern for TCA overdose to Ms. B and her family. Ms. B’s sister reassures the team they will have someone monitor and dispense her medications at home. They decide to discontinue her current psychiatric regimen, and Ms. B is started on nortriptyline 50 mg/d at night, with plans to titrate based on tolerability.</p> <p> <strong>References </strong> </p> <p class="reference"><hl name="2"/> 1. Gaynes BN, Lux L, Gartlehner G, et al. Defining treatment-resistant depression. Depress Anxiety. 2020;37(2):134-145. doi:10.1002/da.22968<br/><br/> 2. Kennedy SH, Lam RW, McIntyre RS, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 clinical guidelines for the management of adults with major depressive disorder: section 3. Pharmacological treatments. Can J Psychiatry. 2016;61(9):540-560. doi:10.1177/0706743716659417<br/><br/> 3. VA/DoD clinical practice guideline for the management of major depressive disorder. Veterans Health Administration and Department of Defense; 2016. https://www.healthquality.va.gov/guidelines/MH/mdd/VADoDMDDCPGFINAL82916.pdf<br/><br/> 4. Gelenberg AJ, Freeman MP, Markowitz JC, et al. Practice guideline for the treatment of patients with major depressive disorder. Am J Psychiatry. 2010;167(Suppl 10):9-118.<br/><br/> 5. Meyer JM. A concise guide to monoamine oxidase inhibitors. Current Psychiatry. 2017;16(12):14-16,18-23,47,A.<br/><br/> 6. Taylor D. Selective serotonin reuptake inhibitors and tricyclic antidepressants in combination. Interactions and therapeutic uses. Br J Psychiatry. 1995;167(5):575-580. doi:10.1192/bjp.167.5.575<br/><br/> 7. Thase ME, Trivedi MH, Rush AJ. MAOIs in the contemporary treatment of depression. Neuropsychopharmacology. 1995;12(3):185-219. doi:10.1016/0893-133X(94)00058-8<br/><br/> 8. Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006;163(11):1905-1917. doi:10.1176/ajp.2006.163.11.1905<br/><br/> 9. McGrath PJ, Stewart JW, Fava M, et al. Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: a STAR*D report. Am J Psychiatry. 2006;163(9):1531-1666. doi:10.1176/ajp.2006.163.9.1531<br/><br/> 10. Kim T, Xu C, Amsterdam JD. Relative effectiveness of tricyclic antidepressant versus monoamine oxidase inhibitor monotherapy for treatment-resistant depression. J Affect Disord. 2019;250:199-203. doi:10.1016/j.jad.2019.03.028<br/><br/> 11. Suchting R, Tirumalajaru V, Gareeb R, et al. Revisiting monoamine oxidase inhibitors for the treatment of depressive disorders: a systematic review and network meta-analysis. J Affect Disord. 2021;282:1153-1160. doi:10.1016/j.jad.2021.01.021
<br/><br/> 12. Stahl SM, Felker A. Monoamine oxidase inhibitors: a modern guide to an unrequited class of antidepressants. CNS Spectr. 2008;13(10):855-870. doi:10.1017/s1092852900016965<br/><br/> 13. Chamberlain SR, Baldwin DS. Monoamine oxidase inhibitors (MAOIs) in psychiatric practice: how to use them safely and effectively. CNS Drugs. 2021;35(7):703-716. doi:10.1007/s40263-021-00832-x<br/><br/> 14. Nardil [package insert]. New York, NY: Parke-Davis; 2009.<br/><br/> 15. Marplan [package insert]. Parsippany, NJ: Validus Pharmaceuticals LLC; 2020.<br/><br/> 16. Parnate [package insert]. Saint Michael, Barbados: Concordia Pharmaceuticals; 2015.<br/><br/> 17. Emsam [package insert]. Morgantown, WV: Mylan Specialty LP; 2014.<br/><br/> 18. Shulman KI, Herrmann N, Walker SE. Current place of monoamine oxidase inhibitors in the treatment of depression. CNS Drugs. 2013;27(10):789-797. doi:10.1007/s40263-013-0097-3<br/><br/> 19. Sub Laban T, Saadabadi A. Monoamine oxidase inhibitors (MAOI). StatPearls Publishing; 2023. https://www.ncbi.nlm.nih.gov/books/NBK539848/<br/><br/> 20. Rabkin JG, Quitkin FM, McGrath P, et al. Adverse reactions to monoamine oxidase inhibitors. Part II. Treatment correlates and clinical management. J Clin Psychopharmacol. 1985;5(1):2-9.<br/><br/> 21. Gillman PK. Advances pertaining to the pharmacology and interactions of irreversible nonselective monoamine oxidase inhibitors. J Clin Psychopharmacol. 2011;31(1):66-74. doi:10.1097/JCP.0b013e31820469ea<br/><br/> 22. Sidhu G, Marwaha R. Phenelzine. StatPearls Publishing; 2023. https://www.ncbi.nlm.nih.gov/books/NBK554508/<br/><br/> 23. Frieling H, Bleich S. Tranylcypromine: new perspectives on an “old” drug. Eur Arch Psychiatry Clin Neurosci. 2006;256(5):268-273. doi:10.1007/s00406-006-0660-8 </p> <p class="reference"> 24. Goodnick PJ. Seligiline transdermal system in depression. Expert Opin Pharmacother. 2007;8(1):59-64. doi:10.1517/14656566.8.1.59</p> <p class="reference"> 25. Edinoff AN, Swinford CR, Odisho AS, et al. Clinically relevant drug interactions with monoamine oxidase inhibitors. Health Psychol Res. 2022;10(4):39576. doi:10.52965/001c.39576<br/><br/> 26. Gillman PK. A reassessment of the safety profile of monoamine oxidase inhibitors: elucidating tired old tyramine myths. J Neural Transm (Vienna). 2018;125(11):1707-1717. doi:10.1007/s00702-018-1932-y<br/><br/> 27. Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03<br/><br/> 28. McCabe-Sellers BJ, Staggs CG, Bogle ML. Tyramine in foods and monoamine oxidase inhibitor drugs: a crossroad where medicine, nutrition, pharmacy, and food industry converge. J Food Composit Anal. 2006;19:S58-S65. doi:10.1016/j.jfca.2005.12.008<br/><br/> 29. Keck PE Jr, Vuckovic A, Pope HG Jr, et al. Acute cardiovascular response to monoamine oxidase inhibitors: a prospective assessment. J Clin Psychopharmacol. 1989;9(3):203-206.<br/><br/> 30. Bodkin JA, Dunlop BW. Moving on with monoamine oxidase inhibitors. Focus (Am Psychiatr Publ). 2021;19(1):50-52. doi:10.1176/appi.focus.20200046<br/><br/> 31. Amsterdam JD, Kim TT. Relative effectiveness of monoamine oxidase inhibitor and tricyclic antidepressant combination therapy for treatment-resistant depression. J Clin Psychopharmacol. 2019;39(6):649-652. doi:10.1097/JCP.0000000000001130<br/><br/> 32. Keks N, Hope J, Keogh S. Switching and stopping antidepressants. Aust Prescr. 2016;39(3):76-83. doi:10.18773/austprescr.2016.039<br/><br/> 33. Israel JA. Combining stimulants and monoamine oxidase inhibitors: a reexamination of the literature and a report of a new treatment combination. Prim Care Companion CNS Disord. 2015;17(6):10.4088/PCC.15br01836. doi:10.4088/PCC.15br01836<br/><br/> 34. Clemons WE, Makela E, Young J. Concomitant use of modafinil and tranylcypromine in a patient with narcolepsy: a case report. Sleep Med. 2004;5(5):509-511. doi:10.1016/j.sleep.2004.06.006<br/><br/> 35. Ashton AK. Modafinil augmentation of phenelzine for residual fatigue in dysthymia. Am J Psychiatry. 2004;161(9):1716-1717. doi:10.1176/appi.ajp.161.9.1716-a<br/><br/> 36. O’Donnell JM, Bies RR, Shelton RC. Drug therapy of depression and anxiety disorders. In: Brunton LL, Hilal-Dandan R, Knollmann BC, eds. <span class="iref">Goodman &amp; Gilman’s: The Pharmacological Basis of Therapeutics</span>. 13th ed. McGraw Hill; 2017. Accessed June 4, 2023. https://accessanesthesiology.mhmedical.com/content.aspx?bookid=2189&amp;sectionid=169518711</p> <p class="reference"> 37. Hiemke C, Bergemann N, Clement HW, et al. Consensus guidelines for therapeutic drug monitoring in neuropsychopharmacology: update 2017. Pharmacopsychiatry. 2018;51(1-02):9-62. doi:10.1055/s-0043-116492</p> <p class="reference"> 38. Amitriptyline hydrochloride [package insert]. East Brunswick, NJ: Unichem Pharmaceuticals (USA); 2021.<br/><br/> 39. Clomipramine hydrochloride [package insert]. East Windsor, NJ: Aurobindo Pharma Limited; 2023.<br/><br/> 40. Doxepin hydrochloride capsules, USP [package insert]. Bedminster, NJ: Alembic Pharmaceuticals Inc; 2021. <br/><br/> 41. Imipramine hydrochloride tablet [package insert]. Fairfield, NJ: Leading Pharma LLC USA; 2022.  <br/><br/> 42. Trimipramine maleate [package insert]. Northvale, NJ: Elite Laboratories Inc; 2021. <br/><br/> 43. Amoxapine [package insert]. Parsippany, NJ: Actavis Pharma Inc; 2015.<br/><br/> 44. Desipramine hydrochloride tablets [package insert]. Bedminster, NJ: Alembic Pharmaceuticals Inc; 2023.<br/><br/> 45. Nortriptyline hydrochloride capsules, USP [package insert]. Parsippany, NJ: Teva Pharmaceuticals Inc; 2021.<br/><br/> 46. Protriptyline hydrochloride [package insert]. Bensalem, PA: Sigmapharm Laboratories, LLC; 2023.<br/><br/> 47. Calvo B, García MJ, Pedraz JL, et al. Pharmacokinetics of amoxapine and its active metabolites. Int J Clin Pharmacol Ther Toxicol. 1985;23(4):180-185.<br/><br/> 48. Ziegler VE, Biggs JT, Wylie LT, et al. Protriptyline kinetics. Clin Pharmacol Ther. 1978;23(5):580-584. doi:10.1002/cpt1978235580<br/><br/> 49. Cleare A, Pariante CM, Young AH, et al. Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol. 2015;29(5):459-525. doi:10.1177/0269881115581093<br/><br/> 50. Richelson E. Synaptic effects of antidepressants. J Clin Psychopharmacol. 1996;16(3 Suppl 2):1S-9S. doi:10.1097/00004714-199606002-00001<br/><br/> 51. Vos CF, Aarnoutse RE, Op de Coul MJM, et al. Tricyclic antidepressants for major depressive disorder: a comprehensive evaluation of current practice in the Netherlands. BMC Psychiatry. 2021;21(1):481. doi:10.1186/s12888-021-03490-x<br/><br/> 52. Alvarez W Jr, Pickworth KK. Safety of antidepressant drugs in the patient with cardiac disease: a review of the literature. Pharmacotherapy. 2003;23(6):754-771. doi:10.1592/phco.23.6.754.32185<br/><br/> 53. Dietch JT, Fine M. The effect of nortriptyline in elderly patients with cardiac conduction disease. J Clin Psychiatry. 1990;51(2):65-67.<br/><br/> 54. Valento M, Liebelt EL. Cyclic antidepressants. In: Nelson LS, Howland M, Lewin NA, et al, eds. <span class="iref">Goldfrank’s Toxicologic Emergencies</span>. 9th ed. McGraw Hill; 2011. Accessed June 10, 2023. https://accesspharmacy.mhmedical.com/content.aspx?bookid=2569&amp;sectionid=210274664 <br/><br/> 55. Woolf AD, Erdman AR, Nelson LS, et al. Tricyclic antidepressant poisoning: an evidence-based consensus guideline for out-of-hospital management. Clin Toxicol (Phila). 2007;45(3):203-233. doi:10.1080/15563650701226192<br/><br/> 56. Baldwin DS, Anderson IM, Nutt DJ, et al. Evidence-based pharmacological treatment of anxiety disorders, post-traumatic stress disorder and obsessive-compulsive disorder: a revision of the 2005 guidelines from the British Association for Psychopharmacology. J Psychopharmacol. 2014;28(5):403-439. doi:10.1177/0269881114525674<br/><br/> 57. Moulin DE, Clark AJ, Gilron I, et al. Pharmacological management of chronic neuropathic pain - consensus statement and guidelines from the Canadian Pain Society. Pain Res Manag. 2007;12(1):13-21. doi:10.1155/2007/730785<br/><br/> 58. Finnerup NB, Attal N, Haroutounian S, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015;14(2):162-173. doi:10.1016/S1474-4422(14)70251-0<br/><br/> 59. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev. 2007;(4):CD005454. doi:10.1002/14651858.CD005454.pub2<br/><br/> 60. Burch R. Antidepressants for preventive treatment of migraine. Curr Treat Options Neurol. 2019;21(4):18. doi:10.1007/s11940-019-0557-2<br/><br/> 61. Ailani J, Burch RC, Robbins MS; Board of Directors of the American Headache Society. The American Headache Society consensus statement: update on integrating new migraine treatments into clinical practice. Headache. 2021;61(7):1021-1039. doi:10.1111/head.14153<br/><br/> 62. Bauer M, Pfennig A, Severus E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for biological treatment of unipolar depressive disorders, part 1: update 2013 on the acute and continuation treatment of unipolar depressive disorders. World J Biol Psychiatry. 2013;14(5):334-385. doi:10.3109/15622975.2013.804195<br/><br/> 63. Jelovac A, Kolshus E, McLoughlin DM. Relapse following successful electroconvulsive therapy for major depression: a meta-analysis. Neuropsychopharmacology. 2013;38(12):2467-2474. doi:10.1038/npp.2013.149<br/><br/> 64. Spencer T, Biederman J, Coffey B, et al. A double-blind comparison of desipramine and placebo in children and adolescents with chronic tic disorder and comorbid attention-deficit/hyperactivity disorder. Arch Gen Psychiatry. 2002;59(7):649-656. doi:10.1001/archpsyc.59.7.649<br/><br/> 65. Spencer T, Biederman J, Wilens T, et al. Pharmacotherapy of attention-deficit hyperactivity disorder across the life cycle. J Am Acad Child Adolesc Psychiatry. 1996;35(4):409-432. doi:10.1097/00004583-199604000-00008<br/><br/> 66. Wilens TE, Biederman J, Prince J, et al. Six-week, double-blind, placebo-controlled study of desipramine for adult attention deficit hyperactivity disorder. Am J Psychiatry. 1996;153(9):1147-1153. doi:10.1176/ajp.153.9.1147</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002645.SIG
Disable zoom
Off

Symptoms of psychosis and OCD in a patient with postpartum depression

Article Type
Changed
Fri, 12/01/2023 - 01:15
Display Headline
Symptoms of psychosis and OCD in a patient with postpartum depression

CASE Thoughts of harming baby

Ms. A, age 37, is G4P2, 4 months postpartum, and breastfeeding. She has major depressive disorder (MDD) with peripartum onset, posttraumatic stress disorder, and mild intellectual disability. For years she has been stable on fluoxetine 40 mg/d and prazosin 2 mg/d. Despite recent titration of her medications, at her most recent outpatient appointment Ms. A reports having a depressed mood with frequent crying, insomnia, a lack of desire to bond with her baby, and feelings of shame. She also says she has had auditory hallucinations and thoughts of harming her baby. Ms. A’s outpatient physician makes an urgent request for her to be evaluated at the psychiatric emergency department (ED).

HISTORY Depression and possible auditory hallucinations

Ms. A developed MDD following the birth of her first child, for which her care team initiated fluoxetine at 20 mg/d and titrated it to 40 mg/d,which was effective. At that time, her outpatient physician documented potential psychotic features, including vague descriptions of derogatory auditory hallucinations. However, it was unclear if these auditory hallucinations were more representative of a distressing inner monologue without the quality of an external voice. The team determined that Ms. A was not at acute risk for harm to herself or her baby and was appropriate for outpatient care. Because the nature of these possible auditory hallucinations was mild, nondistressing, and nonthreatening, the treatment team did not initiate an antipsychotic and Ms. A was not hospitalized. She has no history of hypomanic/manic episodes and has never met criteria for a psychotic disorder.

EVALUATION Distressing thoughts and discontinued medications

During the evaluation by psychiatric emergency services, Ms. A reports that 2 weeks after giving birth she experienced a worsening of her depressive symptoms. She says she began hearing voices telling her to harm herself and her baby and describes frequent distressing thoughts, such as stabbing her baby with a knife and running over her baby with a car. Ms. A says she repeatedly wakes up at night to check on her baby’s breathing, overfeeds her baby due to a fear of inadequate nutrition, and notes intermittent feelings of confusion. Afraid of being alone with her infant, Ms. A asks her partner and mother to move in with her. Additionally, she says 2 weeks ago she discontinued all her medications at the suggestion of her partner, who recommended herbal supplements. Ms. A’s initial routine laboratory results are unremarkable and her urine drug screen is negative for all substances.

[polldaddy:13041928]

The authors’ observations

Approximately 85% of birthing parents experience some form of postpartum mood disturbance; 10% to 15% develop more significant symptoms of anxiety or depression.3 The etiology of postpartum illness is multifactorial, and includes psychiatric personal/family history, insomnia, acute and chronic psychosocial stressors, and rapid hormone fluctuations.1 As a result, the postpartum period represents a vulnerable time for birthing parents, particularly those with previously established psychiatric illness.

Ms. A’s initial presentation was concerning for a possible diagnosis of postpartum psychosis vs obsessive-compulsive disorder (OCD) with postpartum onset; other differential diagnoses included MDD with peripartum onset and psychotic features (Table1-6). Ms. A’s subjective clinical history was significant for critical pertinent findings of both OCD with postpartum onset (ie, egodystonic intrusive thoughts, checking behaviors, feelings of shame, and seeking reassurance) and postpartum psychosis (ie, command auditory hallucinations and waxing/waning confusion), which added to diagnostic complexity.

CP02212048_t1.png

Although postpartum psychosis is rare (1 to 2 cases per 1,000 women),5 it is considered a psychiatric emergency because it has significant potential for infanticide, morbidity, and mortality. Most symptoms develop within the first 2 weeks of the postpartum period.2 There are many risk factors for the development of postpartum psychosis; however, in first-time pregnancies, a previous diagnosis of BD I is the single most important risk factor.1 Approximately 20% to 30% of women with BD experience postpartum psychosis.4

For many patients (approximately 56.7%, according to 1 meta-analysis7), postpartum psychosis denotes an episode of BD, representing a more severe form of illness with increased risk of recurrence. Most manic or mixed mood episodes reoccur within the first year removed from the perinatal period. In contrast, for some patients (approximately 43.5% according to the same meta-analysis), the episode denotes “isolated postpartum psychosis.”7 Isolated postpartum psychosis is a psychotic episode that occurs only in the postpartum period with no recurrence of psychosis or recurrence of psychosis exclusive to postpartum periods. If treated, this type of postpartum psychosis has a more favorable prognosis than postpartum psychosis in a patient with BD.7 As such, a BD diagnosis should not be established at the onset of a patient’s first postpartum psychosis presentation. Regardless of type, all presentations of postpartum psychosis are considered a psychiatry emergency.

Continue to: The prevalence of OCD...

 

 

The prevalence of OCD with postpartum onset varies. One study estimated it occurs in 2.43% of cases.4 However, the true prevalence is likely underreported due to feelings of guilt or shame associated with intrusive thoughts, and fear of stigmatization and separation from the baby. Approximately 70.6% of women experiencing OCD with postpartum onset have a comorbid depressive disorder.4

Ms. A’s presentation to the psychiatric ED carried with it diagnostic complexity and uncertainty. Her initial presentation was concerning for elements of both postpartum psychosis and OCD with postpartum onset. After her evaluation in the psychiatric ED, there remained a lack of clear and convincing evidence for a diagnosis of OCD with postpartum onset, which eliminated the possibility of discharging Ms. A with robust safety planning and reinitiation of a selective serotonin reuptake inhibitor.

Additionally, because auditory hallucinations are atypical in OCD, the treatment team remained concerned for a diagnosis of postpartum psychosis, which would warrant hospitalization. With assistance from the institution’s reproductive psychiatrists, the treatment team discussed the importance of inpatient hospitalization for risk mitigation, close observation, and thorough evaluation for greater diagnostic clarity and certainty.

TREATMENT Involuntary hospitalization

The treatment team counsels Ms. A and her partner on her differential diagnoses, including the elevated acute risk of harm to herself and her baby if she has postpartum psychosis, as well as the need for continued observation and evaluation. When alone with a clinician, Ms. A says she understands and agrees to voluntary hospitalization. However, following a subsequent risk-benefit discussion with her partner, they both grew increasingly concerned about her separation from the baby and reinitiating her medications. Amid these concerns, the treatment team notices that Ms. A attempts to minimize her symptoms. Ms. A changes her mind and no longer consents to hospitalization. She is placed on a psychiatric hold for involuntary hospitalization on the psychiatric inpatient unit.

On the inpatient unit, the inpatient clinicians and a reproductive psychiatrist continue to evaluate Ms. A. Though her diagnosis remains unclear, Ms. A agrees to start a trial of quetiapine 100 mg/d titrated to 150 mg/d to manage her potential postpartum psychosis, depressed mood, insomnia (off-label), anxiety (off-label), and OCD (off-label). Lithium is deferred because Ms. A is breastfeeding.

[polldaddy:13041932]

Continue to: The authors' observations

 

 

The authors’ observations

Due to an elevated acute risk of suicide and infanticide, postpartum psychosis represents a psychiatric emergency and often requires hospitalization. The Figure outlines steps in evaluating a patient with concerns for postpartum psychosis in a psychiatric emergency service setting. Due to the waxing and waning nature of symptoms, patients may appear psychiatrically stable at any time but remain at an overall elevated acute risk of harm to self and/or their baby.

CP02212048_f1.png

If a patient is being considered for discharge based on yes answers to all questions in Step 2 of the Figure, the emergency psychiatric clinician must initiate appropriate psychotropic medications and complete robust safety planning with the patient and a trusted adult who will provide direct supervision. Safety planning may include (but is not limited to) strict return precautions, education on concerning symptoms and behaviors, psychotropic education and agreement of compliance, and detailed instructions on outpatient follow-up within 1 week. Ideally—and as was the case for Ms. A—a reproductive psychiatrist should be consulted in the emergency setting for shared decision-making on admission vs discharge, medication management, and outpatient follow-up considerations.

Because postpartum psychosis carries significant risks and hospitalization generally results in separating the patient from their baby, initiating psychotropics should not be delayed. Clinicians must consider the patient’s psychiatric history, allergies, and breastfeeding status.

Based on current evidence, first-line treatment for postpartum psychosis includes a mood stabilizer, an antipsychotic, and possibly a benzodiazepine.6 Thus, an appropriate initial treatment regimen would be a benzodiazepine (particularly lorazepam due to its relatively shorter half-life) and an antipsychotic (eg, haloperidol, olanzapine, or quetiapine) for acute psychosis, plus lithium for mood stabilization.1,5

If the postpartum psychosis represents an episode of BD, use of a long-term mood stabilizer may be required. In contrast, for isolated postpartum psychosis, clinicians may consider initiating psychotropics only in the immediate postpartum period, with an eventual slow taper. In future pregnancies, psychotropics may be reintroduced postpartum, which will avoid peripartum fetal exposure.8 If the patient is breastfeeding, lithium may be deferred in an acute care setting. For patients with evidence of catatonia, severe suicidality, refusal of oral intake with compromised nutrition, severe agitation, or treatment resistance, electro­convulsive therapy remains a safe and effective treatment option.6 Additionally, the safety of continued breastfeeding in acute psychosis must be considered, with the potential for recommending discontinuation, which would decrease sleep disruptions at night and increase the ability of others to feed the baby. Comprehensive care requires nonpharmacologic interventions, including psychoeducation for the patient and their family, individual psychotherapy, and expansion of psychosocial supports.

Continue to: Patients who have experienced...

 

 

Patients who have experienced an episode of postpartum psychosis are predisposed to another episode in future pregnancies.1 Current research recommends prophylaxis of recurrence with lithium monotherapy.1,2,5,6 Similar to other psychotropics in reproductive psychiatry, maintenance therapy on lithium requires a thorough “risk vs risk” discussion with the patient. The risk of lithium use while pregnant and/or breastfeeding must be weighed against the risks associated with postpartum psychosis (ie, infanticide, suicide, poor peripartum care, or poor infant bonding).

OUTCOME Improved mood

After 7 days of inpatient treatment with quetiapine, Ms. A demonstrates improvement in the targeted depressive symptoms (including improved motivation/energy and insomnia, decreased feelings of guilt, and denial of ongoing suicidal ideation). Additionally, the thoughts of harming her baby are less frequent, and command auditory hallucinations resolve. Upon discharge, Ms. A and her partner meet with inpatient clinicians for continued counseling, safety planning, and plans for outpatient follow-up with the institution’s reproductive psychiatrist.

The authors’ observations

Many aspects of Ms. A’s initial presentation in the psychiatric ED were challenging. Given the presence of symptoms of both psychosis and OCD, a diagnosis was difficult to ascertain in the emergency setting. Since command auditory hallucinations are atypical in patients with postpartum OCD, the treatment team maintained high suspicion for postpartum psychosis, which represented an emergency requiring inpatient care.

Hospitalization separated Ms. A from her baby, for whom she was the primary caregiver. Additional considerations for inpatient admission and psychotropic initiation were necessary, because Ms. A was breastfeeding. Although Ms. A’s partner was able to provide full-time childcare, the patient ultimately did not agree to hospitalization and required an emergency hold for involuntary admission, which was an additional barrier to care. Furthermore, her partner held unfavorable beliefs regarding psychotropic medications and Ms. A’s need for hospital admission, which required ongoing patient and partner education in the emergency, inpatient, and outpatient settings. Moreover, if Ms. A’s symptoms were ultimately attributable to postpartum OCD, the patient’s involuntary hospitalization might have increased the risk of stigmatization of mental illness and treatment with psychotropics.

Bottom Line

The peripartum period is a vulnerable time for patients, particularly those with previously diagnosed psychiatric illnesses. Postpartum psychosis is the most severe form of postpartum psychiatric illness and often represents an episode of bipolar disorder. Due to an elevated acute risk of suicide and infanticide, postpartum psychosis is a psychiatric emergency and warrants inpatient hospitalization for immediate intervention.

Related Resources

Drug Brand Names

Fluoxetine • Prozac
Haloperidol • Haldol
Lithium • Eskalith, Lithobid
Lorazepam • Ativan
Olanzapine • Zyprexa
Prazosin • Minipress
Quetiapine • Seroquel
Sertraline • Zoloft
Valproic acid • Depakene

[embed:render:related:node:199890]

[embed:render:related:node:197703]

References

1. Raza SK, Raza S. Postpartum Psychosis. StatPearls Publishing; 2023. Updated June 26, 2023. https://www.ncbi.nlm.nih.gov/books/NBK544304/

2. MGH Center for Women’s Mental Health. What Is Postpartum Psychosis: This Is What You Need to Know. MGH Center for Women’s Mental Health. Published November 15, 2019. Accessed June 22, 2023. https://womensmentalhealth.org/posts/postpartum-psychosis-ten-things-need-know-2/

3. MGH Center for Women’s Mental Health. Postpartum Psychiatric Disorders. MGH Center for Women’s Mental Health. Accessed October 7, 2023. https://womensmentalhealth.org/specialty-clinics-2/postpartum-psychiatric-disorders-2/

4. Sharma V, Sommerdyk C. Obsessive-compulsive disorder in the postpartum period: diagnosis, differential diagnosis and management. Womens Health (Lond). 2015;11(4):543-552. doi:10.2217/whe.15.20

5. Osborne LM. Recognizing and managing postpartum psychosis: a clinical guide for obstetric providers. Obstet Gynecol Clin North Am. 2018;45(3):455-468. doi:10.1016/j.ogc.2018.04.005

6. Hutner LA, Catapano LA, Nagle-Yang SM, et al, eds. Textbook of Women’s Reproductive Mental Health. American Psychiatric Association; 2022.

7. Gilden J, Kamperman AM, Munk-Olsen T, et al. Long-term outcomes of postpartum psychosis: a systematic review and meta-analysis. J Clin Psychiatry. 2020;81(2):19r12906. doi:10.4088/JCP.19r12906

8. Bergink V, Boyce P, Munk-Olsen T. Postpartum psychosis: a valuable misnomer. Aust N Z J Psychiatry. 2015;49(2):102-103. doi:10.1177/0004867414564698

Article PDF
Author and Disclosure Information

Dr. Rachel Rose is a PGY-2 Psychiatry Resident, Department of Psychiatry, The University of New Mexico, Albuquerque, New Mexico. Dr. Shedd is a Forensic Psychiatry Fellow, Department of Psychiatry, University Hospitals Health System, Case Western Reserve University, Cleveland, Ohio. Drs. Marquette Rose and Higgins are Assistant Professor Reproductive Psychiatrists, Department of Psychiatry, The University of New Mexico, Albuquerque, New Mexico. 

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Issue
Current Psychiatry - 22(12)
Publications
Topics
Page Number
48-53
Sections
Author and Disclosure Information

Dr. Rachel Rose is a PGY-2 Psychiatry Resident, Department of Psychiatry, The University of New Mexico, Albuquerque, New Mexico. Dr. Shedd is a Forensic Psychiatry Fellow, Department of Psychiatry, University Hospitals Health System, Case Western Reserve University, Cleveland, Ohio. Drs. Marquette Rose and Higgins are Assistant Professor Reproductive Psychiatrists, Department of Psychiatry, The University of New Mexico, Albuquerque, New Mexico. 

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Author and Disclosure Information

Dr. Rachel Rose is a PGY-2 Psychiatry Resident, Department of Psychiatry, The University of New Mexico, Albuquerque, New Mexico. Dr. Shedd is a Forensic Psychiatry Fellow, Department of Psychiatry, University Hospitals Health System, Case Western Reserve University, Cleveland, Ohio. Drs. Marquette Rose and Higgins are Assistant Professor Reproductive Psychiatrists, Department of Psychiatry, The University of New Mexico, Albuquerque, New Mexico. 

Disclosures
The authors report no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products.

Article PDF
Article PDF

CASE Thoughts of harming baby

Ms. A, age 37, is G4P2, 4 months postpartum, and breastfeeding. She has major depressive disorder (MDD) with peripartum onset, posttraumatic stress disorder, and mild intellectual disability. For years she has been stable on fluoxetine 40 mg/d and prazosin 2 mg/d. Despite recent titration of her medications, at her most recent outpatient appointment Ms. A reports having a depressed mood with frequent crying, insomnia, a lack of desire to bond with her baby, and feelings of shame. She also says she has had auditory hallucinations and thoughts of harming her baby. Ms. A’s outpatient physician makes an urgent request for her to be evaluated at the psychiatric emergency department (ED).

HISTORY Depression and possible auditory hallucinations

Ms. A developed MDD following the birth of her first child, for which her care team initiated fluoxetine at 20 mg/d and titrated it to 40 mg/d,which was effective. At that time, her outpatient physician documented potential psychotic features, including vague descriptions of derogatory auditory hallucinations. However, it was unclear if these auditory hallucinations were more representative of a distressing inner monologue without the quality of an external voice. The team determined that Ms. A was not at acute risk for harm to herself or her baby and was appropriate for outpatient care. Because the nature of these possible auditory hallucinations was mild, nondistressing, and nonthreatening, the treatment team did not initiate an antipsychotic and Ms. A was not hospitalized. She has no history of hypomanic/manic episodes and has never met criteria for a psychotic disorder.

EVALUATION Distressing thoughts and discontinued medications

During the evaluation by psychiatric emergency services, Ms. A reports that 2 weeks after giving birth she experienced a worsening of her depressive symptoms. She says she began hearing voices telling her to harm herself and her baby and describes frequent distressing thoughts, such as stabbing her baby with a knife and running over her baby with a car. Ms. A says she repeatedly wakes up at night to check on her baby’s breathing, overfeeds her baby due to a fear of inadequate nutrition, and notes intermittent feelings of confusion. Afraid of being alone with her infant, Ms. A asks her partner and mother to move in with her. Additionally, she says 2 weeks ago she discontinued all her medications at the suggestion of her partner, who recommended herbal supplements. Ms. A’s initial routine laboratory results are unremarkable and her urine drug screen is negative for all substances.

[polldaddy:13041928]

The authors’ observations

Approximately 85% of birthing parents experience some form of postpartum mood disturbance; 10% to 15% develop more significant symptoms of anxiety or depression.3 The etiology of postpartum illness is multifactorial, and includes psychiatric personal/family history, insomnia, acute and chronic psychosocial stressors, and rapid hormone fluctuations.1 As a result, the postpartum period represents a vulnerable time for birthing parents, particularly those with previously established psychiatric illness.

Ms. A’s initial presentation was concerning for a possible diagnosis of postpartum psychosis vs obsessive-compulsive disorder (OCD) with postpartum onset; other differential diagnoses included MDD with peripartum onset and psychotic features (Table1-6). Ms. A’s subjective clinical history was significant for critical pertinent findings of both OCD with postpartum onset (ie, egodystonic intrusive thoughts, checking behaviors, feelings of shame, and seeking reassurance) and postpartum psychosis (ie, command auditory hallucinations and waxing/waning confusion), which added to diagnostic complexity.

CP02212048_t1.png

Although postpartum psychosis is rare (1 to 2 cases per 1,000 women),5 it is considered a psychiatric emergency because it has significant potential for infanticide, morbidity, and mortality. Most symptoms develop within the first 2 weeks of the postpartum period.2 There are many risk factors for the development of postpartum psychosis; however, in first-time pregnancies, a previous diagnosis of BD I is the single most important risk factor.1 Approximately 20% to 30% of women with BD experience postpartum psychosis.4

For many patients (approximately 56.7%, according to 1 meta-analysis7), postpartum psychosis denotes an episode of BD, representing a more severe form of illness with increased risk of recurrence. Most manic or mixed mood episodes reoccur within the first year removed from the perinatal period. In contrast, for some patients (approximately 43.5% according to the same meta-analysis), the episode denotes “isolated postpartum psychosis.”7 Isolated postpartum psychosis is a psychotic episode that occurs only in the postpartum period with no recurrence of psychosis or recurrence of psychosis exclusive to postpartum periods. If treated, this type of postpartum psychosis has a more favorable prognosis than postpartum psychosis in a patient with BD.7 As such, a BD diagnosis should not be established at the onset of a patient’s first postpartum psychosis presentation. Regardless of type, all presentations of postpartum psychosis are considered a psychiatry emergency.

Continue to: The prevalence of OCD...

 

 

The prevalence of OCD with postpartum onset varies. One study estimated it occurs in 2.43% of cases.4 However, the true prevalence is likely underreported due to feelings of guilt or shame associated with intrusive thoughts, and fear of stigmatization and separation from the baby. Approximately 70.6% of women experiencing OCD with postpartum onset have a comorbid depressive disorder.4

Ms. A’s presentation to the psychiatric ED carried with it diagnostic complexity and uncertainty. Her initial presentation was concerning for elements of both postpartum psychosis and OCD with postpartum onset. After her evaluation in the psychiatric ED, there remained a lack of clear and convincing evidence for a diagnosis of OCD with postpartum onset, which eliminated the possibility of discharging Ms. A with robust safety planning and reinitiation of a selective serotonin reuptake inhibitor.

Additionally, because auditory hallucinations are atypical in OCD, the treatment team remained concerned for a diagnosis of postpartum psychosis, which would warrant hospitalization. With assistance from the institution’s reproductive psychiatrists, the treatment team discussed the importance of inpatient hospitalization for risk mitigation, close observation, and thorough evaluation for greater diagnostic clarity and certainty.

TREATMENT Involuntary hospitalization

The treatment team counsels Ms. A and her partner on her differential diagnoses, including the elevated acute risk of harm to herself and her baby if she has postpartum psychosis, as well as the need for continued observation and evaluation. When alone with a clinician, Ms. A says she understands and agrees to voluntary hospitalization. However, following a subsequent risk-benefit discussion with her partner, they both grew increasingly concerned about her separation from the baby and reinitiating her medications. Amid these concerns, the treatment team notices that Ms. A attempts to minimize her symptoms. Ms. A changes her mind and no longer consents to hospitalization. She is placed on a psychiatric hold for involuntary hospitalization on the psychiatric inpatient unit.

On the inpatient unit, the inpatient clinicians and a reproductive psychiatrist continue to evaluate Ms. A. Though her diagnosis remains unclear, Ms. A agrees to start a trial of quetiapine 100 mg/d titrated to 150 mg/d to manage her potential postpartum psychosis, depressed mood, insomnia (off-label), anxiety (off-label), and OCD (off-label). Lithium is deferred because Ms. A is breastfeeding.

[polldaddy:13041932]

Continue to: The authors' observations

 

 

The authors’ observations

Due to an elevated acute risk of suicide and infanticide, postpartum psychosis represents a psychiatric emergency and often requires hospitalization. The Figure outlines steps in evaluating a patient with concerns for postpartum psychosis in a psychiatric emergency service setting. Due to the waxing and waning nature of symptoms, patients may appear psychiatrically stable at any time but remain at an overall elevated acute risk of harm to self and/or their baby.

CP02212048_f1.png

If a patient is being considered for discharge based on yes answers to all questions in Step 2 of the Figure, the emergency psychiatric clinician must initiate appropriate psychotropic medications and complete robust safety planning with the patient and a trusted adult who will provide direct supervision. Safety planning may include (but is not limited to) strict return precautions, education on concerning symptoms and behaviors, psychotropic education and agreement of compliance, and detailed instructions on outpatient follow-up within 1 week. Ideally—and as was the case for Ms. A—a reproductive psychiatrist should be consulted in the emergency setting for shared decision-making on admission vs discharge, medication management, and outpatient follow-up considerations.

Because postpartum psychosis carries significant risks and hospitalization generally results in separating the patient from their baby, initiating psychotropics should not be delayed. Clinicians must consider the patient’s psychiatric history, allergies, and breastfeeding status.

Based on current evidence, first-line treatment for postpartum psychosis includes a mood stabilizer, an antipsychotic, and possibly a benzodiazepine.6 Thus, an appropriate initial treatment regimen would be a benzodiazepine (particularly lorazepam due to its relatively shorter half-life) and an antipsychotic (eg, haloperidol, olanzapine, or quetiapine) for acute psychosis, plus lithium for mood stabilization.1,5

If the postpartum psychosis represents an episode of BD, use of a long-term mood stabilizer may be required. In contrast, for isolated postpartum psychosis, clinicians may consider initiating psychotropics only in the immediate postpartum period, with an eventual slow taper. In future pregnancies, psychotropics may be reintroduced postpartum, which will avoid peripartum fetal exposure.8 If the patient is breastfeeding, lithium may be deferred in an acute care setting. For patients with evidence of catatonia, severe suicidality, refusal of oral intake with compromised nutrition, severe agitation, or treatment resistance, electro­convulsive therapy remains a safe and effective treatment option.6 Additionally, the safety of continued breastfeeding in acute psychosis must be considered, with the potential for recommending discontinuation, which would decrease sleep disruptions at night and increase the ability of others to feed the baby. Comprehensive care requires nonpharmacologic interventions, including psychoeducation for the patient and their family, individual psychotherapy, and expansion of psychosocial supports.

Continue to: Patients who have experienced...

 

 

Patients who have experienced an episode of postpartum psychosis are predisposed to another episode in future pregnancies.1 Current research recommends prophylaxis of recurrence with lithium monotherapy.1,2,5,6 Similar to other psychotropics in reproductive psychiatry, maintenance therapy on lithium requires a thorough “risk vs risk” discussion with the patient. The risk of lithium use while pregnant and/or breastfeeding must be weighed against the risks associated with postpartum psychosis (ie, infanticide, suicide, poor peripartum care, or poor infant bonding).

OUTCOME Improved mood

After 7 days of inpatient treatment with quetiapine, Ms. A demonstrates improvement in the targeted depressive symptoms (including improved motivation/energy and insomnia, decreased feelings of guilt, and denial of ongoing suicidal ideation). Additionally, the thoughts of harming her baby are less frequent, and command auditory hallucinations resolve. Upon discharge, Ms. A and her partner meet with inpatient clinicians for continued counseling, safety planning, and plans for outpatient follow-up with the institution’s reproductive psychiatrist.

The authors’ observations

Many aspects of Ms. A’s initial presentation in the psychiatric ED were challenging. Given the presence of symptoms of both psychosis and OCD, a diagnosis was difficult to ascertain in the emergency setting. Since command auditory hallucinations are atypical in patients with postpartum OCD, the treatment team maintained high suspicion for postpartum psychosis, which represented an emergency requiring inpatient care.

Hospitalization separated Ms. A from her baby, for whom she was the primary caregiver. Additional considerations for inpatient admission and psychotropic initiation were necessary, because Ms. A was breastfeeding. Although Ms. A’s partner was able to provide full-time childcare, the patient ultimately did not agree to hospitalization and required an emergency hold for involuntary admission, which was an additional barrier to care. Furthermore, her partner held unfavorable beliefs regarding psychotropic medications and Ms. A’s need for hospital admission, which required ongoing patient and partner education in the emergency, inpatient, and outpatient settings. Moreover, if Ms. A’s symptoms were ultimately attributable to postpartum OCD, the patient’s involuntary hospitalization might have increased the risk of stigmatization of mental illness and treatment with psychotropics.

Bottom Line

The peripartum period is a vulnerable time for patients, particularly those with previously diagnosed psychiatric illnesses. Postpartum psychosis is the most severe form of postpartum psychiatric illness and often represents an episode of bipolar disorder. Due to an elevated acute risk of suicide and infanticide, postpartum psychosis is a psychiatric emergency and warrants inpatient hospitalization for immediate intervention.

Related Resources

Drug Brand Names

Fluoxetine • Prozac
Haloperidol • Haldol
Lithium • Eskalith, Lithobid
Lorazepam • Ativan
Olanzapine • Zyprexa
Prazosin • Minipress
Quetiapine • Seroquel
Sertraline • Zoloft
Valproic acid • Depakene

[embed:render:related:node:199890]

[embed:render:related:node:197703]

CASE Thoughts of harming baby

Ms. A, age 37, is G4P2, 4 months postpartum, and breastfeeding. She has major depressive disorder (MDD) with peripartum onset, posttraumatic stress disorder, and mild intellectual disability. For years she has been stable on fluoxetine 40 mg/d and prazosin 2 mg/d. Despite recent titration of her medications, at her most recent outpatient appointment Ms. A reports having a depressed mood with frequent crying, insomnia, a lack of desire to bond with her baby, and feelings of shame. She also says she has had auditory hallucinations and thoughts of harming her baby. Ms. A’s outpatient physician makes an urgent request for her to be evaluated at the psychiatric emergency department (ED).

HISTORY Depression and possible auditory hallucinations

Ms. A developed MDD following the birth of her first child, for which her care team initiated fluoxetine at 20 mg/d and titrated it to 40 mg/d,which was effective. At that time, her outpatient physician documented potential psychotic features, including vague descriptions of derogatory auditory hallucinations. However, it was unclear if these auditory hallucinations were more representative of a distressing inner monologue without the quality of an external voice. The team determined that Ms. A was not at acute risk for harm to herself or her baby and was appropriate for outpatient care. Because the nature of these possible auditory hallucinations was mild, nondistressing, and nonthreatening, the treatment team did not initiate an antipsychotic and Ms. A was not hospitalized. She has no history of hypomanic/manic episodes and has never met criteria for a psychotic disorder.

EVALUATION Distressing thoughts and discontinued medications

During the evaluation by psychiatric emergency services, Ms. A reports that 2 weeks after giving birth she experienced a worsening of her depressive symptoms. She says she began hearing voices telling her to harm herself and her baby and describes frequent distressing thoughts, such as stabbing her baby with a knife and running over her baby with a car. Ms. A says she repeatedly wakes up at night to check on her baby’s breathing, overfeeds her baby due to a fear of inadequate nutrition, and notes intermittent feelings of confusion. Afraid of being alone with her infant, Ms. A asks her partner and mother to move in with her. Additionally, she says 2 weeks ago she discontinued all her medications at the suggestion of her partner, who recommended herbal supplements. Ms. A’s initial routine laboratory results are unremarkable and her urine drug screen is negative for all substances.

[polldaddy:13041928]

The authors’ observations

Approximately 85% of birthing parents experience some form of postpartum mood disturbance; 10% to 15% develop more significant symptoms of anxiety or depression.3 The etiology of postpartum illness is multifactorial, and includes psychiatric personal/family history, insomnia, acute and chronic psychosocial stressors, and rapid hormone fluctuations.1 As a result, the postpartum period represents a vulnerable time for birthing parents, particularly those with previously established psychiatric illness.

Ms. A’s initial presentation was concerning for a possible diagnosis of postpartum psychosis vs obsessive-compulsive disorder (OCD) with postpartum onset; other differential diagnoses included MDD with peripartum onset and psychotic features (Table1-6). Ms. A’s subjective clinical history was significant for critical pertinent findings of both OCD with postpartum onset (ie, egodystonic intrusive thoughts, checking behaviors, feelings of shame, and seeking reassurance) and postpartum psychosis (ie, command auditory hallucinations and waxing/waning confusion), which added to diagnostic complexity.

CP02212048_t1.png

Although postpartum psychosis is rare (1 to 2 cases per 1,000 women),5 it is considered a psychiatric emergency because it has significant potential for infanticide, morbidity, and mortality. Most symptoms develop within the first 2 weeks of the postpartum period.2 There are many risk factors for the development of postpartum psychosis; however, in first-time pregnancies, a previous diagnosis of BD I is the single most important risk factor.1 Approximately 20% to 30% of women with BD experience postpartum psychosis.4

For many patients (approximately 56.7%, according to 1 meta-analysis7), postpartum psychosis denotes an episode of BD, representing a more severe form of illness with increased risk of recurrence. Most manic or mixed mood episodes reoccur within the first year removed from the perinatal period. In contrast, for some patients (approximately 43.5% according to the same meta-analysis), the episode denotes “isolated postpartum psychosis.”7 Isolated postpartum psychosis is a psychotic episode that occurs only in the postpartum period with no recurrence of psychosis or recurrence of psychosis exclusive to postpartum periods. If treated, this type of postpartum psychosis has a more favorable prognosis than postpartum psychosis in a patient with BD.7 As such, a BD diagnosis should not be established at the onset of a patient’s first postpartum psychosis presentation. Regardless of type, all presentations of postpartum psychosis are considered a psychiatry emergency.

Continue to: The prevalence of OCD...

 

 

The prevalence of OCD with postpartum onset varies. One study estimated it occurs in 2.43% of cases.4 However, the true prevalence is likely underreported due to feelings of guilt or shame associated with intrusive thoughts, and fear of stigmatization and separation from the baby. Approximately 70.6% of women experiencing OCD with postpartum onset have a comorbid depressive disorder.4

Ms. A’s presentation to the psychiatric ED carried with it diagnostic complexity and uncertainty. Her initial presentation was concerning for elements of both postpartum psychosis and OCD with postpartum onset. After her evaluation in the psychiatric ED, there remained a lack of clear and convincing evidence for a diagnosis of OCD with postpartum onset, which eliminated the possibility of discharging Ms. A with robust safety planning and reinitiation of a selective serotonin reuptake inhibitor.

Additionally, because auditory hallucinations are atypical in OCD, the treatment team remained concerned for a diagnosis of postpartum psychosis, which would warrant hospitalization. With assistance from the institution’s reproductive psychiatrists, the treatment team discussed the importance of inpatient hospitalization for risk mitigation, close observation, and thorough evaluation for greater diagnostic clarity and certainty.

TREATMENT Involuntary hospitalization

The treatment team counsels Ms. A and her partner on her differential diagnoses, including the elevated acute risk of harm to herself and her baby if she has postpartum psychosis, as well as the need for continued observation and evaluation. When alone with a clinician, Ms. A says she understands and agrees to voluntary hospitalization. However, following a subsequent risk-benefit discussion with her partner, they both grew increasingly concerned about her separation from the baby and reinitiating her medications. Amid these concerns, the treatment team notices that Ms. A attempts to minimize her symptoms. Ms. A changes her mind and no longer consents to hospitalization. She is placed on a psychiatric hold for involuntary hospitalization on the psychiatric inpatient unit.

On the inpatient unit, the inpatient clinicians and a reproductive psychiatrist continue to evaluate Ms. A. Though her diagnosis remains unclear, Ms. A agrees to start a trial of quetiapine 100 mg/d titrated to 150 mg/d to manage her potential postpartum psychosis, depressed mood, insomnia (off-label), anxiety (off-label), and OCD (off-label). Lithium is deferred because Ms. A is breastfeeding.

[polldaddy:13041932]

Continue to: The authors' observations

 

 

The authors’ observations

Due to an elevated acute risk of suicide and infanticide, postpartum psychosis represents a psychiatric emergency and often requires hospitalization. The Figure outlines steps in evaluating a patient with concerns for postpartum psychosis in a psychiatric emergency service setting. Due to the waxing and waning nature of symptoms, patients may appear psychiatrically stable at any time but remain at an overall elevated acute risk of harm to self and/or their baby.

CP02212048_f1.png

If a patient is being considered for discharge based on yes answers to all questions in Step 2 of the Figure, the emergency psychiatric clinician must initiate appropriate psychotropic medications and complete robust safety planning with the patient and a trusted adult who will provide direct supervision. Safety planning may include (but is not limited to) strict return precautions, education on concerning symptoms and behaviors, psychotropic education and agreement of compliance, and detailed instructions on outpatient follow-up within 1 week. Ideally—and as was the case for Ms. A—a reproductive psychiatrist should be consulted in the emergency setting for shared decision-making on admission vs discharge, medication management, and outpatient follow-up considerations.

Because postpartum psychosis carries significant risks and hospitalization generally results in separating the patient from their baby, initiating psychotropics should not be delayed. Clinicians must consider the patient’s psychiatric history, allergies, and breastfeeding status.

Based on current evidence, first-line treatment for postpartum psychosis includes a mood stabilizer, an antipsychotic, and possibly a benzodiazepine.6 Thus, an appropriate initial treatment regimen would be a benzodiazepine (particularly lorazepam due to its relatively shorter half-life) and an antipsychotic (eg, haloperidol, olanzapine, or quetiapine) for acute psychosis, plus lithium for mood stabilization.1,5

If the postpartum psychosis represents an episode of BD, use of a long-term mood stabilizer may be required. In contrast, for isolated postpartum psychosis, clinicians may consider initiating psychotropics only in the immediate postpartum period, with an eventual slow taper. In future pregnancies, psychotropics may be reintroduced postpartum, which will avoid peripartum fetal exposure.8 If the patient is breastfeeding, lithium may be deferred in an acute care setting. For patients with evidence of catatonia, severe suicidality, refusal of oral intake with compromised nutrition, severe agitation, or treatment resistance, electro­convulsive therapy remains a safe and effective treatment option.6 Additionally, the safety of continued breastfeeding in acute psychosis must be considered, with the potential for recommending discontinuation, which would decrease sleep disruptions at night and increase the ability of others to feed the baby. Comprehensive care requires nonpharmacologic interventions, including psychoeducation for the patient and their family, individual psychotherapy, and expansion of psychosocial supports.

Continue to: Patients who have experienced...

 

 

Patients who have experienced an episode of postpartum psychosis are predisposed to another episode in future pregnancies.1 Current research recommends prophylaxis of recurrence with lithium monotherapy.1,2,5,6 Similar to other psychotropics in reproductive psychiatry, maintenance therapy on lithium requires a thorough “risk vs risk” discussion with the patient. The risk of lithium use while pregnant and/or breastfeeding must be weighed against the risks associated with postpartum psychosis (ie, infanticide, suicide, poor peripartum care, or poor infant bonding).

OUTCOME Improved mood

After 7 days of inpatient treatment with quetiapine, Ms. A demonstrates improvement in the targeted depressive symptoms (including improved motivation/energy and insomnia, decreased feelings of guilt, and denial of ongoing suicidal ideation). Additionally, the thoughts of harming her baby are less frequent, and command auditory hallucinations resolve. Upon discharge, Ms. A and her partner meet with inpatient clinicians for continued counseling, safety planning, and plans for outpatient follow-up with the institution’s reproductive psychiatrist.

The authors’ observations

Many aspects of Ms. A’s initial presentation in the psychiatric ED were challenging. Given the presence of symptoms of both psychosis and OCD, a diagnosis was difficult to ascertain in the emergency setting. Since command auditory hallucinations are atypical in patients with postpartum OCD, the treatment team maintained high suspicion for postpartum psychosis, which represented an emergency requiring inpatient care.

Hospitalization separated Ms. A from her baby, for whom she was the primary caregiver. Additional considerations for inpatient admission and psychotropic initiation were necessary, because Ms. A was breastfeeding. Although Ms. A’s partner was able to provide full-time childcare, the patient ultimately did not agree to hospitalization and required an emergency hold for involuntary admission, which was an additional barrier to care. Furthermore, her partner held unfavorable beliefs regarding psychotropic medications and Ms. A’s need for hospital admission, which required ongoing patient and partner education in the emergency, inpatient, and outpatient settings. Moreover, if Ms. A’s symptoms were ultimately attributable to postpartum OCD, the patient’s involuntary hospitalization might have increased the risk of stigmatization of mental illness and treatment with psychotropics.

Bottom Line

The peripartum period is a vulnerable time for patients, particularly those with previously diagnosed psychiatric illnesses. Postpartum psychosis is the most severe form of postpartum psychiatric illness and often represents an episode of bipolar disorder. Due to an elevated acute risk of suicide and infanticide, postpartum psychosis is a psychiatric emergency and warrants inpatient hospitalization for immediate intervention.

Related Resources

Drug Brand Names

Fluoxetine • Prozac
Haloperidol • Haldol
Lithium • Eskalith, Lithobid
Lorazepam • Ativan
Olanzapine • Zyprexa
Prazosin • Minipress
Quetiapine • Seroquel
Sertraline • Zoloft
Valproic acid • Depakene

[embed:render:related:node:199890]

[embed:render:related:node:197703]

References

1. Raza SK, Raza S. Postpartum Psychosis. StatPearls Publishing; 2023. Updated June 26, 2023. https://www.ncbi.nlm.nih.gov/books/NBK544304/

2. MGH Center for Women’s Mental Health. What Is Postpartum Psychosis: This Is What You Need to Know. MGH Center for Women’s Mental Health. Published November 15, 2019. Accessed June 22, 2023. https://womensmentalhealth.org/posts/postpartum-psychosis-ten-things-need-know-2/

3. MGH Center for Women’s Mental Health. Postpartum Psychiatric Disorders. MGH Center for Women’s Mental Health. Accessed October 7, 2023. https://womensmentalhealth.org/specialty-clinics-2/postpartum-psychiatric-disorders-2/

4. Sharma V, Sommerdyk C. Obsessive-compulsive disorder in the postpartum period: diagnosis, differential diagnosis and management. Womens Health (Lond). 2015;11(4):543-552. doi:10.2217/whe.15.20

5. Osborne LM. Recognizing and managing postpartum psychosis: a clinical guide for obstetric providers. Obstet Gynecol Clin North Am. 2018;45(3):455-468. doi:10.1016/j.ogc.2018.04.005

6. Hutner LA, Catapano LA, Nagle-Yang SM, et al, eds. Textbook of Women’s Reproductive Mental Health. American Psychiatric Association; 2022.

7. Gilden J, Kamperman AM, Munk-Olsen T, et al. Long-term outcomes of postpartum psychosis: a systematic review and meta-analysis. J Clin Psychiatry. 2020;81(2):19r12906. doi:10.4088/JCP.19r12906

8. Bergink V, Boyce P, Munk-Olsen T. Postpartum psychosis: a valuable misnomer. Aust N Z J Psychiatry. 2015;49(2):102-103. doi:10.1177/0004867414564698

References

1. Raza SK, Raza S. Postpartum Psychosis. StatPearls Publishing; 2023. Updated June 26, 2023. https://www.ncbi.nlm.nih.gov/books/NBK544304/

2. MGH Center for Women’s Mental Health. What Is Postpartum Psychosis: This Is What You Need to Know. MGH Center for Women’s Mental Health. Published November 15, 2019. Accessed June 22, 2023. https://womensmentalhealth.org/posts/postpartum-psychosis-ten-things-need-know-2/

3. MGH Center for Women’s Mental Health. Postpartum Psychiatric Disorders. MGH Center for Women’s Mental Health. Accessed October 7, 2023. https://womensmentalhealth.org/specialty-clinics-2/postpartum-psychiatric-disorders-2/

4. Sharma V, Sommerdyk C. Obsessive-compulsive disorder in the postpartum period: diagnosis, differential diagnosis and management. Womens Health (Lond). 2015;11(4):543-552. doi:10.2217/whe.15.20

5. Osborne LM. Recognizing and managing postpartum psychosis: a clinical guide for obstetric providers. Obstet Gynecol Clin North Am. 2018;45(3):455-468. doi:10.1016/j.ogc.2018.04.005

6. Hutner LA, Catapano LA, Nagle-Yang SM, et al, eds. Textbook of Women’s Reproductive Mental Health. American Psychiatric Association; 2022.

7. Gilden J, Kamperman AM, Munk-Olsen T, et al. Long-term outcomes of postpartum psychosis: a systematic review and meta-analysis. J Clin Psychiatry. 2020;81(2):19r12906. doi:10.4088/JCP.19r12906

8. Bergink V, Boyce P, Munk-Olsen T. Postpartum psychosis: a valuable misnomer. Aust N Z J Psychiatry. 2015;49(2):102-103. doi:10.1177/0004867414564698

Issue
Current Psychiatry - 22(12)
Issue
Current Psychiatry - 22(12)
Page Number
48-53
Page Number
48-53
Publications
Publications
Topics
Article Type
Display Headline
Symptoms of psychosis and OCD in a patient with postpartum depression
Display Headline
Symptoms of psychosis and OCD in a patient with postpartum depression
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>1123 Cases Rose</fileName> <TBEID>0C02E573.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02E573</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Symptoms of psychosis and OCD in</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CP</TBLocation> <QCDate/> <firstPublished>20231129T083452</firstPublished> <LastPublished>20231129T083452</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231129T083451</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Rachel Rose, MD, Megan Shedd, MD, Marquette Rose, MD</byline> <bylineText/> <bylineFull>Rachel Rose, MD, Megan Shedd, MD, Marquette Rose, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>48-53</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Ms. A, age 37, is G4P2, 4 months postpartum, and breastfeeding. She has major depressive disorder (MDD) with peripartum onset, posttraumatic stress disorder, an</metaDescription> <articlePDF>299302</articlePDF> <teaserImage/> <title>Symptoms of psychosis and OCD in a patient with postpartum depression</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>December</pubPubdateMonth> <pubPubdateDay/> <pubVolume>22</pubVolume> <pubNumber>12</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>1985</CMSID> </CMSIDs> <keywords> <keyword>schizophrenia</keyword> <keyword> obsessive-compulsive disorder</keyword> <keyword> depression</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CP</publicationCode> <pubIssueName>December 2023</pubIssueName> <pubArticleType>Audio | 1985</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Current Psychiatry</journalTitle> <journalFullTitle>Current Psychiatry</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">11</term> </publications> <sections> <term canonical="true">47</term> </sections> <topics> <term canonical="true">293</term> <term>202</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002647.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Symptoms of psychosis and OCD in a patient with postpartum depression</title> <deck/> </itemMeta> <itemContent> <p> <strong>Rachel Rose, MD, Megan Shedd, MD, Marquette Rose, MD, and Nina Higgins, MD</strong> </p> <h3><span class="dingbat"> </span><span class="box">CASE</span><span class="dingbat"> </span> Thoughts of harming baby</h3> <p>Ms. A, age 37, is G4P2, 4 months postpartum, and breastfeeding. She has major depressive disorder (MDD) with peripartum onset, posttraumatic stress disorder, and mild intellectual disability. For years she has been stable on fluoxetine 40 mg/d and prazosin 2 mg/d. Despite recent titration of her medications, at her most recent outpatient appointment Ms. A reports having a depressed mood with frequent crying, insomnia, a lack of desire to bond with her baby, and feelings of shame. She also says she has had auditory hallucinations and thoughts of harming her baby. Ms. A’s outpatient physician makes an urgent request for her to be evaluated at the psychiatric emergency department (ED).</p> <h3><span class="dingbat"> </span><span class="box">HISTORY</span><span class="dingbat"> </span> Depression and possible auditory hallucinations</h3> <p>Ms. A developed MDD following the birth of her first child, for which her care team initiated fluoxetine at 20 mg/d and titrated it to 40 mg/d,<strong/>which was effective. At that time, her outpatient physician documented potential psychotic features, including vague descriptions of derogatory auditory hallucinations. However, it was unclear if these auditory hallucinations were more representative of a distressing inner monologue without the quality of an external voice. The team determined that Ms. A was not at acute risk for harm to herself or her baby and was appropriate for outpatient care. Because the nature of these possible auditory hallucinations was mild, nondistressing, and nonthreatening, the treatment team did not initiate an antipsychotic and Ms. A was not hospitalized. She has no history of hypomanic/manic episodes and has never met criteria for a psychotic disorder.</p> <h3><span class="dingbat"> </span><span class="box">EVALUATION</span><span class="dingbat"> </span> Distressing thoughts and discontinued medications</h3> <p>During the evaluation by psychiatric emergency services, Ms. A reports that 2 weeks after giving birth she experienced a worsening of her depressive symptoms. She says she began hearing voices telling her to harm herself and her baby and describes frequent distressing thoughts, such as stabbing her baby with a knife and running over her baby with a car. Ms. A says she repeatedly wakes up at night to check on her baby’s breathing, overfeeds her baby due to a fear of inadequate nutrition, and notes intermittent feelings of confusion. Afraid of being alone with her infant, Ms. A asks her partner and mother to move in with her. Additionally, she says 2 weeks ago she discontinued all her medications at the suggestion of her partner, who recommended herbal supplements. Ms. A’s initial routine laboratory results are unremarkable and her urine drug screen is negative for all substances.</p> <p> <strong>What is the most robust risk factor in the development of postpartum psychosis?<sup>1,2</sup></strong> </p> <p>a) History of schizophrenia<br/><br/>b) History of bipolar disorder (BD)<br/><br/>c) Substance use disorder during pregnancy<br/><br/>d) History of MDD with psychotic features</p> <h3>The authors’ <span class="observation">observations</span></h3> <p>Approximately 85% of birthing parents experience some form of postpartum mood disturbance; 10% to 15% develop more significant symptoms of anxiety or depression.<sup>3</sup> The etiology of postpartum illness is multifactorial, and includes psychiatric personal/family history, insomnia, acute and chronic psychosocial stressors, and rapid hormone fluctuations.<sup>1</sup> As a result, the postpartum period represents a vulnerable time for birthing parents, particularly those with previously established psychiatric illness.</p> <p>Ms. A’s initial presentation was concerning for a possible diagnosis of postpartum psychosis vs obsessive-compulsive disorder (OCD) with postpartum onset; other differential diagnoses included MDD with peripartum onset and psychotic features (<b><i>Table</i></b>,<sup>1-6</sup> <b><i>page 49</i></b>). Ms. A’s subjective clinical history was significant for critical pertinent findings of both OCD with postpartum onset (ie, egodystonic intrusive thoughts, checking behaviors, feelings of shame, and seeking reassurance) and postpartum psychosis (ie, command auditory hallucinations and waxing/waning confusion), which added to diagnostic complexity.<br/><br/>Although postpartum psychosis is rare (1 to 2 cases per 1,000 women),<sup>5</sup> it is considered a psychiatric emergency because it has significant potential for infanticide, morbidity, and mortality. Most symptoms develop within the first 2 weeks of the postpartum period.<sup>2</sup> There are many risk factors for the development of postpartum psychosis; however, in first-time pregnancies, a previous diagnosis of BD I is the single most important risk factor.<sup>1</sup> Approximately 20% to 30% of women with BD experience postpartum psychosis.<sup>4<br/><br/></sup>For many patients (approximately 56.7%, according to 1 meta-analysis<sup>7</sup>), postpartum psychosis denotes an episode of BD, representing a more severe form of illness with increased risk of recurrence. Most manic or mixed mood episodes reoccur within the first year removed from the perinatal period. In contrast, for some patients (approximately 43.5% according to the same meta-analysis), the episode denotes “isolated postpartum psychosis.”<sup>7</sup> Isolated postpartum psychosis is a psychotic episode that occurs only in the postpartum period with no recurrence of psychosis or recurrence of psychosis exclusive to postpartum periods. If treated, this type of postpartum psychosis has a more favorable prognosis than postpartum psychosis in a patient with BD.<sup>7</sup> As such, a BD diagnosis should not be established at the onset of a patient’s first postpartum psychosis presentation. Regardless of type, all presentations of postpartum psychosis are considered a psychiatry emergency.<br/><br/>The prevalence of OCD with postpartum onset varies. One study estimated it occurs in 2.43% of cases.<sup>4</sup> However, the true prevalence is likely underreported due to feelings of guilt or shame associated with intrusive thoughts, and fear of stigmatization and separation from the baby. Approximately 70.6% of women experiencing OCD with postpartum onset have a comorbid depressive disorder.<sup>4<br/><br/></sup>Ms. A’s presentation to the psychiatric ED carried with it diagnostic complexity and uncertainty. Her initial presentation was concerning for elements of both postpartum psychosis and OCD with postpartum onset. After her evaluation in the psychiatric ED, there remained a lack of clear and convincing evidence for a diagnosis of OCD with postpartum onset, which eliminated the possibility of discharging Ms. A with robust safety planning and reinitiation of a selective serotonin reuptake inhibitor.<br/><br/>Additionally, because auditory hallucinations are atypical in OCD, the treatment team remained concerned for a diagnosis of postpartum psychosis, which would warrant hospitalization. With assistance from the institution’s reproductive psychiatrists, the treatment team discussed the importance of inpatient hospitalization for risk mitigation, close observation, and thorough evaluation for greater diagnostic clarity and certainty.</p> <h3><span class="dingbat"> </span><span class="box">TREATMENT</span><span class="dingbat"> </span> Involuntary hospitalization</h3> <p>The treatment team counsels Ms. A and her partner on her differential diagnoses, including the elevated acute risk of harm to herself and her baby if she has postpartum psychosis, as well as the need for continued observation and evaluation. When alone with a clinician, Ms. A says she understands and agrees to voluntary hospitalization. However, following a subsequent risk-benefit discussion with her partner, they both grew increasingly concerned about her separation from the baby and reinitiating her medications. Amid these concerns, the treatment team notices that Ms. A attempts to minimize her symptoms. Ms. A changes her mind and no longer consents to hospitalization. She is placed on a psychiatric hold for involuntary hospitalization on the psychiatric inpatient unit.</p> <p>On the inpatient unit, the inpatient clinicians and a reproductive psychiatrist continue to evaluate Ms. A. Though her diagnosis remains unclear, Ms. A agrees to start a trial of quetiapine 100 mg/d titrated to 150 mg/d to manage her potential postpartum psychosis, depressed mood, insomnia (off-label), anxiety (off-label), and OCD (off-label). Lithium is deferred because Ms. A is breastfeeding.</p> <p> <strong>Ms. A is planning another pregnancy.To prevent future episodes of postpartum psychosis, which psychotropic would you recommend?<sup>1,5,7</sup></strong> </p> <p>a) Haloperidol<br/><br/>b) Valproic acid<br/><br/>c) Lithium or a second-generation antipsychotic<br/><br/>d) Sertraline</p> <h3>The authors’ <span class="observation">observations</span></h3> <p>Due to an elevated acute risk of suicide and infanticide, postpartum psychosis represents a psychiatric emergency and often requires hospitalization. The <span class="bitalic">Figure</span> (<span class="bitalic">page 52</span>) outlines steps in evaluating a patient with concerns for postpartum psychosis in a psychiatric emergency service setting. Due to the waxing and waning nature of symptoms, patients may appear psychiatrically stable at any time but remain at an overall elevated acute risk of harm to self and/or their baby.</p> <p>If a patient is being considered for discharge based on yes answers to all questions in Step 2 of the <b><i>Figure</i></b>, the emergency psychiatric clinician must initiate appropriate psychotropic medications and complete robust safety planning with the patient and a trusted adult who will provide direct supervision. Safety planning may include (but is not limited to) strict return precautions, education on concerning symptoms and behaviors, psychotropic education and agreement of compliance, and detailed instructions on outpatient follow-up within 1 week. Ideally—and as was the case for Ms. A—a reproductive psychiatrist should be consulted in the emergency setting for shared decision-making on admission vs discharge, medication management, and outpatient follow-up considerations.<br/><br/>Because postpartum psychosis carries significant risks and hospitalization generally results in separating the patient from their baby, initiating psychotropics should not be delayed. Clinicians must consider the patient’s psychiatric history, allergies, and breastfeeding status.<br/><br/>Based on current evidence, first-line treatment for postpartum psychosis includes a mood stabilizer, an antipsychotic, and possibly a benzodiazepine.<sup>6</sup> Thus, an appropriate initial treatment regimen would be a benzodiazepine (particularly lorazepam due to its relatively shorter half-life) and an antipsychotic (eg, haloperidol, olanzapine, or quetiapine) for acute psychosis, plus lithium for mood stabilization.<sup>1,5<br/><br/></sup>If the postpartum psychosis represents an episode of BD, use of a long-term mood stabilizer may be required. In contrast, for isolated postpartum psychosis, clinicians may consider initiating psychotropics only in the immediate postpartum period, with an eventual slow taper. In future pregnancies, psychotropics may be reintroduced postpartum, which will avoid peripartum fetal exposure.<sup>8</sup> If the patient is breastfeeding, lithium may be deferred in an acute care setting. For patients with evidence of catatonia, severe suicidality, refusal of oral intake with compromised nutrition, severe agitation, or treatment resistance, electro­convulsive therapy remains a safe and effective treatment option.<sup>6</sup> Additionally, the safety of continued breastfeeding in acute psychosis must be considered, with the potential for recommending discontinuation, which would decrease sleep disruptions at night and increase the ability of others to feed the baby. Comprehensive care requires nonpharmacologic interventions, including psychoeducation for the patient and their family, individual psychotherapy, and expansion of psychosocial supports.<br/><br/>Patients who have experienced an episode of postpartum psychosis are predisposed to another episode in future pregnancies.<sup>1</sup> Current research recommends prophylaxis of recurrence with lithium monotherapy.<sup>1,2,5,6</sup> Similar to other psychotropics in reproductive psychiatry, maintenance therapy on lithium requires a thorough “risk vs risk” discussion with the patient. The risk of lithium use while pregnant and/or breastfeeding must be weighed against the risks associated with postpartum psychosis (ie, infanticide, suicide, poor peripartum care, or poor infant bonding).</p> <h3><span class="dingbat"> </span><span class="box">OUTCOME</span><span class="dingbat"> </span> Improved mood</h3> <p>After 7 days of inpatient treatment with quetiapine, Ms. A demonstrates improvement in the targeted depressive symptoms (including improved motivation/energy and insomnia, decreased feelings of guilt, and denial of ongoing suicidal ideation). Additionally, the thoughts of harming her baby are less frequent, and command auditory hallucinations resolve. Upon discharge, Ms. A and her partner meet with inpatient clinicians for continued counseling, safety planning, and plans for outpatient follow-up with the institution’s reproductive psychiatrist.</p> <h3>The authors’ <span class="observation">observations</span></h3> <p>Many aspects of Ms. A’s initial presentation in the psychiatric ED were challenging. Given the presence of symptoms of both psychosis and OCD, a diagnosis was difficult to ascertain in the emergency setting. Since command auditory hallucinations are atypical in patients with postpartum OCD, the treatment team maintained high suspicion for postpartum psychosis, which represented an emergency requiring inpatient care.</p> <p>Hospitalization separated Ms. A from her baby, for whom she was the primary caregiver. Additional considerations for inpatient admission and psychotropic initiation were necessary, because Ms. A was breastfeeding. Although Ms. A’s partner was able to provide full-time childcare, the patient ultimately did not agree to hospitalization and required an emergency hold for involuntary admission, which was an additional barrier to care. Furthermore, her partner held unfavorable beliefs regarding psychotropic medications and Ms. A’s need for hospital admission, which required ongoing patient and partner education in the emergency, inpatient, and outpatient settings. Moreover, if Ms. A’s symptoms were ultimately attributable to postpartum OCD, the patient’s involuntary hospitalization might have increased the risk of stigmatization of mental illness and treatment with psychotropics.</p> <p> <strong>References</strong> </p> <p class="reference"> 1. Raza SK, Raza S. <i>Postpartum Psychosis</i>. StatPearls Publishing; 2023. Updated June 26, 2023. https://www.ncbi.nlm.nih.gov/books/NBK544304/<br/><br/> 2. MGH Center for Women’s Mental Health. What Is Postpartum Psychosis: This Is What You Need to Know. MGH Center for Women’s Mental Health. Published November 15, 2019. Accessed June 22, 2023. https://womensmentalhealth.org/posts/postpartum-psychosis-ten-things-need-know-2/<br/><br/> 3. MGH Center for Women’s Mental Health. Postpartum Psychiatric Disorders. MGH Center for Women’s Mental Health. Accessed October 7, 2023. https://womensmentalhealth.org/specialty-clinics-2/postpartum-psychiatric-disorders-2/<br/><br/> 4. Sharma V, Sommerdyk C. Obsessive-compulsive disorder in the postpartum period: diagnosis, differential diagnosis and management. Womens Health (Lond). 2015;11(4):543-552. doi:10.2217/whe.15.20<br/><br/> 5. Osborne LM. Recognizing and managing postpartum psychosis: a clinical guide for obstetric providers. Obstet Gynecol Clin North Am. 2018;45(3):455-468. doi:10.1016/j.ogc.2018.04.005<br/><br/> 6. Hutner LA, Catapano LA, Nagle-Yang SM, et al, eds. <span class="iref">Textbook of Women’s Reproductive Mental Health.</span> American Psychiatric Association; 2022.<br/><br/> 7. Gilden J, Kamperman AM, Munk-Olsen T, et al. Long-term outcomes of postpartum psychosis: a systematic review and meta-analysis. J Clin Psychiatry. 2020;81(2):19r12906. doi:10.4088/JCP.19r12906<br/><br/> 8. Bergink V, Boyce P, Munk-Olsen T. Postpartum psychosis: a valuable misnomer. Aust N Z J Psychiatry. 2015;49(2):102-103. doi:10.1177/0004867414564698</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18002647.SIG
Disable zoom
Off