National Initiative to Prevent Suicide: A new proposal to improve the understanding and prevention of suicide

Article Type
Changed
Mon, 04/16/2018 - 14:38
Display Headline
National Initiative to Prevent Suicide: A new proposal to improve the understanding and prevention of suicide

Suicide is a staggering, tragic, and growing cause of death in the United States. One out of every 62 Americans will die from suicide, based on the national lifetime prevalence rate.1 More than 42,000 Americans died from suicide in 2014, making suicide the second leading cause of death in individuals age 15 to 34, the fourth leading cause among those age 35 to 54, and the tenth leading cause of death in the country overall.2 The incidence of suicide in the general population of the United States increased by 24% between 1999 and 2014.3 This tragedy obviously is not solving itself.

The proposal

U.S. Centers for Disease Control and Prevention (CDC) publishes statistics about the number of suicides, as well as demographic information, collected from coroners and medical examiners across the country. However, these sources do not provide a biological sample that could be used to gather data concerning DNA, RNA, and other potential blood markers, including those reflecting inflammatory and epigenetic processes. However, such biological samples are commonly collected by the U.S. medicolegal death investigation system. In 2003, this system investigated 450,000 unnatural and/or unexplained deaths (ie, approximately 20% of the 2.4 million deaths in the United States that year).4

Each unnatural or unexplained death is examined, often extensively, by a coroner or medical examiner. This examination system costs more than $600 million annually. Yet the data that are collected are handled on a case-by-case and often county-by-county basis, rather than in aggregate. The essence of the proposal presented here is to take the information and biological samples collected in this process and put them into a National Suicide Database (NSD), which then can serve as a resource for scientists to increase our understanding of the genetic, epigenetic, and other factors underlying death due to suicide. This increased understanding will result in the development more effective tools to detect to those at risk for suicide (ie, risk factor tests), to monitor treatment, and to develop new treatments based on a better understanding of the underlying pathophysiology and pathogenesis of suicide. These tools will reduce:

  • the number of lives lost to suicide
  • the pain and suffering of loved ones
  • lost productivity to society, especially when one considers that suicide disproportionately affects individuals during the most productive period of their lives (ie, age 15 to 54).

The NSD will be organized as a government–private partnership, with the government represented by the National Institutes of Health (NIH) and/or the CDC. The goal will be to take the information that is currently being collected by the nation’s medicolegal death investigation system, including the biological samples, systematize it, enter it into a common database, and make it available to qualified researchers across the country. The administrative arm of the system will be responsible for ensuring systematic data collection, storage in a searchable and integrated database housed within the NIH and/or the CDC, and vetting researchers who will have access to the data, including those with expertise in genomics, molecular biology, suicide, epidemiology, and data-mining. (Currently, the CDC’s National Violent Death Reporting System, which is a state-based surveillance system, pools data on violent deaths from multiple sources into a usable, anonymous database. These sources include state and local medical examiners, coroners, law enforcement, crime labs, and vital statistics records, but they do not include any biological material even though it is collected [personal correspondence with the CDC, July 2016].)

Because information on suicides currently are handled primarily on a county-by-county basis, data concerning these deaths are not facilitating a better understanding of the causes and strategies for preventing suicide. Correcting this situation is the goal of this proposal, as modeled by the National Cancer Institute’s War on Cancer, which has transformed the treatment and the outcomes of cancer. If this proposal is enacted, the same type of transformation will occur and result in a reduction in the suicide rate and better outcomes for the psychiatric illnesses that underlie most instances of suicide.

The proposed NSD will address a major and common problem for researchers in this area—small sample sizes. When considered from the perspective of the size of samples feasible for most independent research teams to collect and study, suicide on an annual basis is rare—however, that is not the case when the incidence of suicide in the nation as a whole is considered. In contrast to the data concerning suicides that individual research teams can collect, the proposed genomic database will grow by approximately 40,000 individuals every year, until a meaningful reduction in deaths due to suicide is achieved.

From a research perspective, suicide, although tragic, is one of the few binary outcomes in psychiatry—that is, life or death. Although there may be >1 genetic and/or epigenetic contributor to suicide, within a relatively short period of time, the proposed database will amass—and continue to amass on an ongoing basis—data from a large population of suicide victims. Researchers then can compare the findings from this database with the normative human genome, looking for variants that are over-represented in the population of those who have died by suicide.

Environmental factors undoubtedly also contribute to the risk of suicide, given that the incidence of suicide increases with age, particularly among white males, and with the addition of psychiatric and medical comorbidities. Inflammatory processes also have been implicated in the pathophysiology of a number of psychiatric disorders, including major depression, which is the primary psychiatric risk factor for suicide. Therefore, consideration should be given to collecting whole blood samples if the time between death and autopsy is within an appropriate limit to obtain interpretable data concerning RNA (ie, gene expression) and even biomarkers of inflammatory and other processes at the time of the suicide. This approach has been used by Niculescu et al5,6 for whole blood gene expression. The rationale for using samples of whole blood is that this strategy could be more easily adapted to clinical practice in contrast to using samples from the target organ (ie, brain) or cerebrospinal fluid.

 

 

Roadblocks to progress. In the absence of this proposed NSD, progress in this area has been stymied despite concerted governmental efforts (Box7-10). One reason for the lack of progress has been that governmental efforts have focused on a public health model rather than also including a basic science model aimed at exploring the biological mechanisms underlying the risk of death from suicide. In the current decentralized system, individual researchers and even teams of researchers cannot easily collect data from a sufficiently large population of suicide victims to make inroads in gaining the needed understanding.

Because of the relatively small samples that individual research teams can collect in a reasonable period of time (ie, in terms of grant cycles), many investigators have studied suicide attempts as a surrogate for suicide itself, undoubtedly because suicide attempts are more numerous than suicides themselves, making it easier to collect data. However, there is evidence that these 2 populations—suicide attempters vs those who die by suicide—only partially overlap.

First, the frequency of suicide attempts is 10 to 20 times higher than actual suicides. Second, suicide attempters are 3 times more likely to be female whereas those who die by suicide are 4 times more likely to be male. Third, most individuals who die by suicide do so on their first or second attempt, whereas individuals who have made ≥4 attempts have an increased risk of future attempts rather than for completed suicide compared with the general population. Fourth, certain psychiatric illnesses are more often associated with death by suicide (particularly major depressive disorder, bipolar disorder, and schizophrenia in the first 5 years of an illness) whereas multiple suicide attempts are more often associated with other psychiatric diagnoses such as antisocial and borderline personality disorders.

Finally, in a study in men with a psychiatric disorder, Niculescu et al5 started with 412 candidate genes and found that 208 were associated with suicidal ideation but not suicide itself, whereas 76 genes were associated with both suicidal ideation and completion. Taken together, this evidence suggests that findings concerning suicide attempters, especially those who have made multiple (ie, >3) attempts, might not be extrapolatable to the population of actual suicides.

Is there evidence that this proposal could work?

Yes, research supports the potential utility of the proposed NSD, and this section highlights some of the major findings from these studies, although this review is not intended to be exhaustive.

First, considerable evidence exists for a biological basis for the risk of death due to suicide. The concordance rates for suicide are 10 times higher in monozygotic (“identical”) vs dizygotic (“fraternal”) twins (24.1% vs 2.8%) and 2 to 5 times higher in relatives of those who die by suicide than in the general population. Heritability estimates of fatal suicides and nonfatal suicide attempts in biological relatives of adoptees who die from suicide range from 17% to 45%.11

Second, studies using information from small samples that was arduously collected by individual research groups have yielded important positive data. Most recently, in 2015, a multidisciplinary group led by Niculescu et al5 at Indiana University and other institutions described a test that could predict suicidality in men. This test was developed on the basis of a within-participant discovery approach to identify genes that change in expression between states of no suicidal ideation and high suicidal ideation, which was combined with clinical information assessed by 2 scales, the Convergent Functional Information for Suicidality and the Simplified Affective State Scale. Gene expression was measured in whole blood collected postmortem unless the method of suicide involved a medication overdose that could affect gene expression. These researchers identified 76 genes that likely were involved in suicidal ideation and suicide.

This report had a number of limitations.5 All of the individuals in these studies were being treated for psychiatric illness, were being closely followed by the investigators, and all were male. In addition, as noted above, suicides by overdose were eliminated from the analysis.

In a subsequent study published in 2016, the Niculescu group6 extended their work to women and identified 50 genes contributing to suicide risk in women. Underscoring the need for larger samples, only 3 of the top contributing genes were seen in both men and women, suggesting that there are likely significant sex differences in the biology of suicide completion. This important work needs to be replicated and extended.

In addition to these remarkable advances made in genetic understanding of the risk of suicide, recent research also has demonstrated a role for epigenetic and inflammatory processes as contributors to suicide risk.12-15

There are likely many contributors, including genetic, epigenetic, and environmental factors such as inflammatory processes, that increase the risk of suicide. The goal of this article is not to provide an exhaustive or integrative review of research in this area but rather to argue for the establishment of a national initiative to study all of these factors and to begin that process by establishing the NSD.

 

 

What will be the foreseeable outcome of this initiative?

The establishment of the NSD is expected to lead to better identification of those who are genetically at increased risk of suicide as well as biological factors (eg, inflammatory or other processes) and environmental factors (eg, drug abuse), which can turn that genetic risk into reality. Using research results made possible by the implementation of this proposal, objective testing can be developed to monitor risk more effectively than is currently possible using clinical assessment alone.

Furthermore, this work also can provide targets for developing new treatments. For example, there is convergence between the work of Niculescu et al,5,6 who identified genetic biomarkers for mechanistic target of rapamycin (mTOR) signaling as a risk factor in individuals who died by suicide and the work of Li et al and other researchers,16-18 whose findings have implicated mTOR-dependent synapse formation as a mechanism underlying the rapid (ie, within hours to a couple of days) antidepressant effects of N-methyl-d-aspartate antagonists, such as ketamine, CP-101,606, and esketamine. In fact, the authors of a study presented earlier this year reported that esketamine—an active enantiomer of ketamine—rapidly reduced suicidal ideation as well as other depressive symptoms in individuals admitted to the hospital for suicidal ideation.19 (mTOR is a serine/threonine protein kinase that regulates a number of biological processes in addition to synaptogenesis, including cell growth, cell proliferation, cell motility, cell survival, protein synthesis, and autophagy.20,21)

In aggregate, establishment of this proposed database will facilitate identification of biological (and therefore pharmaceutical) mechanisms beyond those involving biogenic amines, which have been the exclusive biological targets for antidepressants for the past 50 years.22 The likely consequences of the findings generated from research made possible by the proposed NSD will open completely new vistas for helping people at risk for suicide and psychiatric illnesses.

What foreseeable obstacles will need to be addressed?

Of course, obstacles and problems will arise but these will not exceed those encountered by the War on Cancer and they can similarly be overcome with sufficient public support and cooperation. Potential obstacles include:

  • need for incremental funding
  • obtaining the cooperation of the offices of each county medical examiner or coroner in a process that includes uniform systematic data collection
  • determining the situations (eg, time after death and means of death) that will allow for meaningful collection of data such as RNA and inflammatory biomarkers
  • establishing how data and particularly biological samples will be transported and stored
  • issues related to privacy of health information particularly for relatives of suicide victims
  • ensuring the reliability, validity, and comparability of the data received from different medical examiners and coroners.

With regard to the last issue, because stigma is associated with death by suicide, some true suicides could be missed, which would compromise sensitivity but simultaneously increase specificity. Other obstacles or problems may arise; however, I am certain that all such issues are surmountable and that the resulting NSD will be much better than what we have now and will propel our understanding of the biological underpinnings of the loss of life to suicide. (The author proposed a similar but even more ambitious plan 25 years ago,23 but he believes that this is an idea whose time has come.)

Acknowledgments

The author thanks Wayne C. Drevets, MD, Alexander Niculescu, MD, PhD, John Oldman, MD, and John Savitz, PhD, David Sheehan, MD, and Matthew Macaluso, DO for their review and suggestions concerning this proposal/manuscript, and Kaylee Hervey, MPH, from the Sedgwick County Health Department, Wichita, Kansas, for her input. The author also thanks Ruth Ross, as always, for her excellent editing and general assistance.

References

1. Pompili M, Gonda X, Serafini G, et al. Epidemiology of suicide in bipolar disorders: a systematic review of the literature. Bipolar Disord. 2013;15(5):457-490.
2. National Vital Statistics System; National Center for Health Statistics; Centers for Disease Control and Prevention. Ten leading causes of death by age group, United States–2014. Centers for Disease Control and Prevention. http://www.cdc.gov/injury/images/lc-charts/leading_causes_of_death_age_group_2014_1050w760h.gif. Accessed October 17, 2016.
3. Curtin SC, Warner M, Hedegaard H, et al. Increase in suicide in the United States, 1999-2014. National Center for Health Statistics Data Brief No. 241. Atlanta GA: National Center for Health Statistics, U.S. Department of Health and Human Services. http://www.cdc.gov/nchs/products/databriefs/db241.htm. Published April 2016. Accessed June 30, 2016.
4. Committee for the Workshop on the Medicolegal Death Investigation System; Board on Health Promotion and Disease Prevention. Medicolegal death investigation system: workshop summary. Washington, DC: National Academies Press; 2003.
5. Niculescu AB, Levey DF, Phalen PL, et al. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol Psychiatry. 2015;20(11):1266-1285.
6. Levey DF, Niculescu EM, Le-Niculescu H, et al. Towards understanding and predicting suicidality in women: biomarkers and clinical risk assessment. Mol Psychiatry. 2016;21(6):768-785.
7. World Health Organization. Prevention of suicide: guidelines for the formulation and implementation of national strategies. Geneva, Switzerland: World Health Organization; 1996.
8. U.S. Public Health Service. The Surgeon General’s call to action to prevent suicide. Washington, DC: U.S. Public Health Service; 1999.
9. U.S. Department of Health and Human Services (HHS). National Strategy for Suicide Prevention: goals and objectives for action. Rockville, MD: U.S. Department of Health and Human Services; 2001.
10. U.S. Department of Health and Human Services (HHS). National Strategy for Suicide Prevention: goals and objectives for action. Rockville, MD; U.S. Department of Health and Human Services; 2012.
11. Brent DA, Melham N. Familial transmission of suicidal behavior. Psychiatr Clin North Am. 2008;31(2):157-177.
12. Guintivano J, Brown T, Newcomer A, et al. Identification and replication of a combined epigenetic and genetic biomarker predicting suicide and suicidal behaviors. Am J Psychiatry. 2014;171(12):1287-1296.
13. Bay-Richter C, Linderholm KR, Lim CK, et al. A role for inflammatory metabolites as modulators of the glutamate N-methyl-D-aspartate receptor in depression and suicidality. Brain Behav Immun. 2015;43:110-117.
14. Brundin L, Bryleva EY, Thirtamara Rajamani K. Role of inflammation in suicide: from mechanisms to treatment [published online July 27, 2016]. Neuropsychopharmacology. doi: 10.1038/npp.2016.116.
15. Steiner J, Walter M, Gos T, et al. Severe depression is associated with increased microglial quinolinic acid in subregions of the anterior cingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission? J Neuroinflammation. 2011;8:94.
16. Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329(5994):959-964.
17. Zarate CA Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856-864.
18. Preskorn SH, Baker B, Kolluri S, et al. An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-D-aspartate antagonist, CP-101,606, in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol. 2008;28(6):631-637.
19. Canuso C, Singh J, Fedgchin M, et al. PeRSEVERe: a study of esketamine for the rapid reduction of the symptoms of major depressive disorder, including suicidal ideation, in subjects assessed to be at imminent risk for suicide. Presentation at the Annual Meeting of the American Society of Clinical Psychopharmacology, Scottsdale AZ, May 30-June 3, 2016.
20. Brown EJ, Albers MW, Shin TB, et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature. 1994;369(6483):756-758.
21. Moore PA, Rosen CA, Carter KC. Assignment of the human FKBP12-rapamycin-associated protein (FRAP) gene to chromosome 1p36 by fluorescence in situ hybridization. Genomics. 1996;33(2):331-332.
22. Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev. 2004;18(16):1926-1945.
23. Preskorn SH. The future and psychopharmacology: potentials and needs. Psychiatr Ann. 1990;20(11):625-633.

Article PDF
Author and Disclosure Information

Sheldon H. Preskorn, MD

Professor
Department of Psychiatry
University of Kansas School of Medicine-Wichita
Wichita, Kansas
Research Professor
Laureate Institute for Brain Research
Tulsa, Oklahoma

Dr. Preskorn is a Section Editor on the Current Psychiatry Editorial Board

Disclosure

Over the past year, Dr. Preskorn has received grant/research support from or has served as a consultant, on the advisory board, or on the speakers bureau for Acadia, Alkermes, Assurex Health, BioXcel, Eisai, FDA Office of Clinical Pharmacology, Forum, Janssen, Merck, Neurocrine, the National Institute of Mental Health, Naurex/Aptinyx, Rugen Holdings, Salamandra on behalf of Sun Pharmaceuticals, The Stanley Medical Research Institute, Sunovion, and Vanda.

Adapted with permission from: Preskorn SH. National Initiative to Prevent Suicide (NIPS): a new proposal to improve the understanding and prevention of suicide. J Psychiatr Pract. 2016;22(5):398-404.

Issue
November 2016
Publications
Topics
Page Number
24,30-32,36-37
Sections
Author and Disclosure Information

Sheldon H. Preskorn, MD

Professor
Department of Psychiatry
University of Kansas School of Medicine-Wichita
Wichita, Kansas
Research Professor
Laureate Institute for Brain Research
Tulsa, Oklahoma

Dr. Preskorn is a Section Editor on the Current Psychiatry Editorial Board

Disclosure

Over the past year, Dr. Preskorn has received grant/research support from or has served as a consultant, on the advisory board, or on the speakers bureau for Acadia, Alkermes, Assurex Health, BioXcel, Eisai, FDA Office of Clinical Pharmacology, Forum, Janssen, Merck, Neurocrine, the National Institute of Mental Health, Naurex/Aptinyx, Rugen Holdings, Salamandra on behalf of Sun Pharmaceuticals, The Stanley Medical Research Institute, Sunovion, and Vanda.

Adapted with permission from: Preskorn SH. National Initiative to Prevent Suicide (NIPS): a new proposal to improve the understanding and prevention of suicide. J Psychiatr Pract. 2016;22(5):398-404.

Author and Disclosure Information

Sheldon H. Preskorn, MD

Professor
Department of Psychiatry
University of Kansas School of Medicine-Wichita
Wichita, Kansas
Research Professor
Laureate Institute for Brain Research
Tulsa, Oklahoma

Dr. Preskorn is a Section Editor on the Current Psychiatry Editorial Board

Disclosure

Over the past year, Dr. Preskorn has received grant/research support from or has served as a consultant, on the advisory board, or on the speakers bureau for Acadia, Alkermes, Assurex Health, BioXcel, Eisai, FDA Office of Clinical Pharmacology, Forum, Janssen, Merck, Neurocrine, the National Institute of Mental Health, Naurex/Aptinyx, Rugen Holdings, Salamandra on behalf of Sun Pharmaceuticals, The Stanley Medical Research Institute, Sunovion, and Vanda.

Adapted with permission from: Preskorn SH. National Initiative to Prevent Suicide (NIPS): a new proposal to improve the understanding and prevention of suicide. J Psychiatr Pract. 2016;22(5):398-404.

Article PDF
Article PDF

Suicide is a staggering, tragic, and growing cause of death in the United States. One out of every 62 Americans will die from suicide, based on the national lifetime prevalence rate.1 More than 42,000 Americans died from suicide in 2014, making suicide the second leading cause of death in individuals age 15 to 34, the fourth leading cause among those age 35 to 54, and the tenth leading cause of death in the country overall.2 The incidence of suicide in the general population of the United States increased by 24% between 1999 and 2014.3 This tragedy obviously is not solving itself.

The proposal

U.S. Centers for Disease Control and Prevention (CDC) publishes statistics about the number of suicides, as well as demographic information, collected from coroners and medical examiners across the country. However, these sources do not provide a biological sample that could be used to gather data concerning DNA, RNA, and other potential blood markers, including those reflecting inflammatory and epigenetic processes. However, such biological samples are commonly collected by the U.S. medicolegal death investigation system. In 2003, this system investigated 450,000 unnatural and/or unexplained deaths (ie, approximately 20% of the 2.4 million deaths in the United States that year).4

Each unnatural or unexplained death is examined, often extensively, by a coroner or medical examiner. This examination system costs more than $600 million annually. Yet the data that are collected are handled on a case-by-case and often county-by-county basis, rather than in aggregate. The essence of the proposal presented here is to take the information and biological samples collected in this process and put them into a National Suicide Database (NSD), which then can serve as a resource for scientists to increase our understanding of the genetic, epigenetic, and other factors underlying death due to suicide. This increased understanding will result in the development more effective tools to detect to those at risk for suicide (ie, risk factor tests), to monitor treatment, and to develop new treatments based on a better understanding of the underlying pathophysiology and pathogenesis of suicide. These tools will reduce:

  • the number of lives lost to suicide
  • the pain and suffering of loved ones
  • lost productivity to society, especially when one considers that suicide disproportionately affects individuals during the most productive period of their lives (ie, age 15 to 54).

The NSD will be organized as a government–private partnership, with the government represented by the National Institutes of Health (NIH) and/or the CDC. The goal will be to take the information that is currently being collected by the nation’s medicolegal death investigation system, including the biological samples, systematize it, enter it into a common database, and make it available to qualified researchers across the country. The administrative arm of the system will be responsible for ensuring systematic data collection, storage in a searchable and integrated database housed within the NIH and/or the CDC, and vetting researchers who will have access to the data, including those with expertise in genomics, molecular biology, suicide, epidemiology, and data-mining. (Currently, the CDC’s National Violent Death Reporting System, which is a state-based surveillance system, pools data on violent deaths from multiple sources into a usable, anonymous database. These sources include state and local medical examiners, coroners, law enforcement, crime labs, and vital statistics records, but they do not include any biological material even though it is collected [personal correspondence with the CDC, July 2016].)

Because information on suicides currently are handled primarily on a county-by-county basis, data concerning these deaths are not facilitating a better understanding of the causes and strategies for preventing suicide. Correcting this situation is the goal of this proposal, as modeled by the National Cancer Institute’s War on Cancer, which has transformed the treatment and the outcomes of cancer. If this proposal is enacted, the same type of transformation will occur and result in a reduction in the suicide rate and better outcomes for the psychiatric illnesses that underlie most instances of suicide.

The proposed NSD will address a major and common problem for researchers in this area—small sample sizes. When considered from the perspective of the size of samples feasible for most independent research teams to collect and study, suicide on an annual basis is rare—however, that is not the case when the incidence of suicide in the nation as a whole is considered. In contrast to the data concerning suicides that individual research teams can collect, the proposed genomic database will grow by approximately 40,000 individuals every year, until a meaningful reduction in deaths due to suicide is achieved.

From a research perspective, suicide, although tragic, is one of the few binary outcomes in psychiatry—that is, life or death. Although there may be >1 genetic and/or epigenetic contributor to suicide, within a relatively short period of time, the proposed database will amass—and continue to amass on an ongoing basis—data from a large population of suicide victims. Researchers then can compare the findings from this database with the normative human genome, looking for variants that are over-represented in the population of those who have died by suicide.

Environmental factors undoubtedly also contribute to the risk of suicide, given that the incidence of suicide increases with age, particularly among white males, and with the addition of psychiatric and medical comorbidities. Inflammatory processes also have been implicated in the pathophysiology of a number of psychiatric disorders, including major depression, which is the primary psychiatric risk factor for suicide. Therefore, consideration should be given to collecting whole blood samples if the time between death and autopsy is within an appropriate limit to obtain interpretable data concerning RNA (ie, gene expression) and even biomarkers of inflammatory and other processes at the time of the suicide. This approach has been used by Niculescu et al5,6 for whole blood gene expression. The rationale for using samples of whole blood is that this strategy could be more easily adapted to clinical practice in contrast to using samples from the target organ (ie, brain) or cerebrospinal fluid.

 

 

Roadblocks to progress. In the absence of this proposed NSD, progress in this area has been stymied despite concerted governmental efforts (Box7-10). One reason for the lack of progress has been that governmental efforts have focused on a public health model rather than also including a basic science model aimed at exploring the biological mechanisms underlying the risk of death from suicide. In the current decentralized system, individual researchers and even teams of researchers cannot easily collect data from a sufficiently large population of suicide victims to make inroads in gaining the needed understanding.

Because of the relatively small samples that individual research teams can collect in a reasonable period of time (ie, in terms of grant cycles), many investigators have studied suicide attempts as a surrogate for suicide itself, undoubtedly because suicide attempts are more numerous than suicides themselves, making it easier to collect data. However, there is evidence that these 2 populations—suicide attempters vs those who die by suicide—only partially overlap.

First, the frequency of suicide attempts is 10 to 20 times higher than actual suicides. Second, suicide attempters are 3 times more likely to be female whereas those who die by suicide are 4 times more likely to be male. Third, most individuals who die by suicide do so on their first or second attempt, whereas individuals who have made ≥4 attempts have an increased risk of future attempts rather than for completed suicide compared with the general population. Fourth, certain psychiatric illnesses are more often associated with death by suicide (particularly major depressive disorder, bipolar disorder, and schizophrenia in the first 5 years of an illness) whereas multiple suicide attempts are more often associated with other psychiatric diagnoses such as antisocial and borderline personality disorders.

Finally, in a study in men with a psychiatric disorder, Niculescu et al5 started with 412 candidate genes and found that 208 were associated with suicidal ideation but not suicide itself, whereas 76 genes were associated with both suicidal ideation and completion. Taken together, this evidence suggests that findings concerning suicide attempters, especially those who have made multiple (ie, >3) attempts, might not be extrapolatable to the population of actual suicides.

Is there evidence that this proposal could work?

Yes, research supports the potential utility of the proposed NSD, and this section highlights some of the major findings from these studies, although this review is not intended to be exhaustive.

First, considerable evidence exists for a biological basis for the risk of death due to suicide. The concordance rates for suicide are 10 times higher in monozygotic (“identical”) vs dizygotic (“fraternal”) twins (24.1% vs 2.8%) and 2 to 5 times higher in relatives of those who die by suicide than in the general population. Heritability estimates of fatal suicides and nonfatal suicide attempts in biological relatives of adoptees who die from suicide range from 17% to 45%.11

Second, studies using information from small samples that was arduously collected by individual research groups have yielded important positive data. Most recently, in 2015, a multidisciplinary group led by Niculescu et al5 at Indiana University and other institutions described a test that could predict suicidality in men. This test was developed on the basis of a within-participant discovery approach to identify genes that change in expression between states of no suicidal ideation and high suicidal ideation, which was combined with clinical information assessed by 2 scales, the Convergent Functional Information for Suicidality and the Simplified Affective State Scale. Gene expression was measured in whole blood collected postmortem unless the method of suicide involved a medication overdose that could affect gene expression. These researchers identified 76 genes that likely were involved in suicidal ideation and suicide.

This report had a number of limitations.5 All of the individuals in these studies were being treated for psychiatric illness, were being closely followed by the investigators, and all were male. In addition, as noted above, suicides by overdose were eliminated from the analysis.

In a subsequent study published in 2016, the Niculescu group6 extended their work to women and identified 50 genes contributing to suicide risk in women. Underscoring the need for larger samples, only 3 of the top contributing genes were seen in both men and women, suggesting that there are likely significant sex differences in the biology of suicide completion. This important work needs to be replicated and extended.

In addition to these remarkable advances made in genetic understanding of the risk of suicide, recent research also has demonstrated a role for epigenetic and inflammatory processes as contributors to suicide risk.12-15

There are likely many contributors, including genetic, epigenetic, and environmental factors such as inflammatory processes, that increase the risk of suicide. The goal of this article is not to provide an exhaustive or integrative review of research in this area but rather to argue for the establishment of a national initiative to study all of these factors and to begin that process by establishing the NSD.

 

 

What will be the foreseeable outcome of this initiative?

The establishment of the NSD is expected to lead to better identification of those who are genetically at increased risk of suicide as well as biological factors (eg, inflammatory or other processes) and environmental factors (eg, drug abuse), which can turn that genetic risk into reality. Using research results made possible by the implementation of this proposal, objective testing can be developed to monitor risk more effectively than is currently possible using clinical assessment alone.

Furthermore, this work also can provide targets for developing new treatments. For example, there is convergence between the work of Niculescu et al,5,6 who identified genetic biomarkers for mechanistic target of rapamycin (mTOR) signaling as a risk factor in individuals who died by suicide and the work of Li et al and other researchers,16-18 whose findings have implicated mTOR-dependent synapse formation as a mechanism underlying the rapid (ie, within hours to a couple of days) antidepressant effects of N-methyl-d-aspartate antagonists, such as ketamine, CP-101,606, and esketamine. In fact, the authors of a study presented earlier this year reported that esketamine—an active enantiomer of ketamine—rapidly reduced suicidal ideation as well as other depressive symptoms in individuals admitted to the hospital for suicidal ideation.19 (mTOR is a serine/threonine protein kinase that regulates a number of biological processes in addition to synaptogenesis, including cell growth, cell proliferation, cell motility, cell survival, protein synthesis, and autophagy.20,21)

In aggregate, establishment of this proposed database will facilitate identification of biological (and therefore pharmaceutical) mechanisms beyond those involving biogenic amines, which have been the exclusive biological targets for antidepressants for the past 50 years.22 The likely consequences of the findings generated from research made possible by the proposed NSD will open completely new vistas for helping people at risk for suicide and psychiatric illnesses.

What foreseeable obstacles will need to be addressed?

Of course, obstacles and problems will arise but these will not exceed those encountered by the War on Cancer and they can similarly be overcome with sufficient public support and cooperation. Potential obstacles include:

  • need for incremental funding
  • obtaining the cooperation of the offices of each county medical examiner or coroner in a process that includes uniform systematic data collection
  • determining the situations (eg, time after death and means of death) that will allow for meaningful collection of data such as RNA and inflammatory biomarkers
  • establishing how data and particularly biological samples will be transported and stored
  • issues related to privacy of health information particularly for relatives of suicide victims
  • ensuring the reliability, validity, and comparability of the data received from different medical examiners and coroners.

With regard to the last issue, because stigma is associated with death by suicide, some true suicides could be missed, which would compromise sensitivity but simultaneously increase specificity. Other obstacles or problems may arise; however, I am certain that all such issues are surmountable and that the resulting NSD will be much better than what we have now and will propel our understanding of the biological underpinnings of the loss of life to suicide. (The author proposed a similar but even more ambitious plan 25 years ago,23 but he believes that this is an idea whose time has come.)

Acknowledgments

The author thanks Wayne C. Drevets, MD, Alexander Niculescu, MD, PhD, John Oldman, MD, and John Savitz, PhD, David Sheehan, MD, and Matthew Macaluso, DO for their review and suggestions concerning this proposal/manuscript, and Kaylee Hervey, MPH, from the Sedgwick County Health Department, Wichita, Kansas, for her input. The author also thanks Ruth Ross, as always, for her excellent editing and general assistance.

Suicide is a staggering, tragic, and growing cause of death in the United States. One out of every 62 Americans will die from suicide, based on the national lifetime prevalence rate.1 More than 42,000 Americans died from suicide in 2014, making suicide the second leading cause of death in individuals age 15 to 34, the fourth leading cause among those age 35 to 54, and the tenth leading cause of death in the country overall.2 The incidence of suicide in the general population of the United States increased by 24% between 1999 and 2014.3 This tragedy obviously is not solving itself.

The proposal

U.S. Centers for Disease Control and Prevention (CDC) publishes statistics about the number of suicides, as well as demographic information, collected from coroners and medical examiners across the country. However, these sources do not provide a biological sample that could be used to gather data concerning DNA, RNA, and other potential blood markers, including those reflecting inflammatory and epigenetic processes. However, such biological samples are commonly collected by the U.S. medicolegal death investigation system. In 2003, this system investigated 450,000 unnatural and/or unexplained deaths (ie, approximately 20% of the 2.4 million deaths in the United States that year).4

Each unnatural or unexplained death is examined, often extensively, by a coroner or medical examiner. This examination system costs more than $600 million annually. Yet the data that are collected are handled on a case-by-case and often county-by-county basis, rather than in aggregate. The essence of the proposal presented here is to take the information and biological samples collected in this process and put them into a National Suicide Database (NSD), which then can serve as a resource for scientists to increase our understanding of the genetic, epigenetic, and other factors underlying death due to suicide. This increased understanding will result in the development more effective tools to detect to those at risk for suicide (ie, risk factor tests), to monitor treatment, and to develop new treatments based on a better understanding of the underlying pathophysiology and pathogenesis of suicide. These tools will reduce:

  • the number of lives lost to suicide
  • the pain and suffering of loved ones
  • lost productivity to society, especially when one considers that suicide disproportionately affects individuals during the most productive period of their lives (ie, age 15 to 54).

The NSD will be organized as a government–private partnership, with the government represented by the National Institutes of Health (NIH) and/or the CDC. The goal will be to take the information that is currently being collected by the nation’s medicolegal death investigation system, including the biological samples, systematize it, enter it into a common database, and make it available to qualified researchers across the country. The administrative arm of the system will be responsible for ensuring systematic data collection, storage in a searchable and integrated database housed within the NIH and/or the CDC, and vetting researchers who will have access to the data, including those with expertise in genomics, molecular biology, suicide, epidemiology, and data-mining. (Currently, the CDC’s National Violent Death Reporting System, which is a state-based surveillance system, pools data on violent deaths from multiple sources into a usable, anonymous database. These sources include state and local medical examiners, coroners, law enforcement, crime labs, and vital statistics records, but they do not include any biological material even though it is collected [personal correspondence with the CDC, July 2016].)

Because information on suicides currently are handled primarily on a county-by-county basis, data concerning these deaths are not facilitating a better understanding of the causes and strategies for preventing suicide. Correcting this situation is the goal of this proposal, as modeled by the National Cancer Institute’s War on Cancer, which has transformed the treatment and the outcomes of cancer. If this proposal is enacted, the same type of transformation will occur and result in a reduction in the suicide rate and better outcomes for the psychiatric illnesses that underlie most instances of suicide.

The proposed NSD will address a major and common problem for researchers in this area—small sample sizes. When considered from the perspective of the size of samples feasible for most independent research teams to collect and study, suicide on an annual basis is rare—however, that is not the case when the incidence of suicide in the nation as a whole is considered. In contrast to the data concerning suicides that individual research teams can collect, the proposed genomic database will grow by approximately 40,000 individuals every year, until a meaningful reduction in deaths due to suicide is achieved.

From a research perspective, suicide, although tragic, is one of the few binary outcomes in psychiatry—that is, life or death. Although there may be >1 genetic and/or epigenetic contributor to suicide, within a relatively short period of time, the proposed database will amass—and continue to amass on an ongoing basis—data from a large population of suicide victims. Researchers then can compare the findings from this database with the normative human genome, looking for variants that are over-represented in the population of those who have died by suicide.

Environmental factors undoubtedly also contribute to the risk of suicide, given that the incidence of suicide increases with age, particularly among white males, and with the addition of psychiatric and medical comorbidities. Inflammatory processes also have been implicated in the pathophysiology of a number of psychiatric disorders, including major depression, which is the primary psychiatric risk factor for suicide. Therefore, consideration should be given to collecting whole blood samples if the time between death and autopsy is within an appropriate limit to obtain interpretable data concerning RNA (ie, gene expression) and even biomarkers of inflammatory and other processes at the time of the suicide. This approach has been used by Niculescu et al5,6 for whole blood gene expression. The rationale for using samples of whole blood is that this strategy could be more easily adapted to clinical practice in contrast to using samples from the target organ (ie, brain) or cerebrospinal fluid.

 

 

Roadblocks to progress. In the absence of this proposed NSD, progress in this area has been stymied despite concerted governmental efforts (Box7-10). One reason for the lack of progress has been that governmental efforts have focused on a public health model rather than also including a basic science model aimed at exploring the biological mechanisms underlying the risk of death from suicide. In the current decentralized system, individual researchers and even teams of researchers cannot easily collect data from a sufficiently large population of suicide victims to make inroads in gaining the needed understanding.

Because of the relatively small samples that individual research teams can collect in a reasonable period of time (ie, in terms of grant cycles), many investigators have studied suicide attempts as a surrogate for suicide itself, undoubtedly because suicide attempts are more numerous than suicides themselves, making it easier to collect data. However, there is evidence that these 2 populations—suicide attempters vs those who die by suicide—only partially overlap.

First, the frequency of suicide attempts is 10 to 20 times higher than actual suicides. Second, suicide attempters are 3 times more likely to be female whereas those who die by suicide are 4 times more likely to be male. Third, most individuals who die by suicide do so on their first or second attempt, whereas individuals who have made ≥4 attempts have an increased risk of future attempts rather than for completed suicide compared with the general population. Fourth, certain psychiatric illnesses are more often associated with death by suicide (particularly major depressive disorder, bipolar disorder, and schizophrenia in the first 5 years of an illness) whereas multiple suicide attempts are more often associated with other psychiatric diagnoses such as antisocial and borderline personality disorders.

Finally, in a study in men with a psychiatric disorder, Niculescu et al5 started with 412 candidate genes and found that 208 were associated with suicidal ideation but not suicide itself, whereas 76 genes were associated with both suicidal ideation and completion. Taken together, this evidence suggests that findings concerning suicide attempters, especially those who have made multiple (ie, >3) attempts, might not be extrapolatable to the population of actual suicides.

Is there evidence that this proposal could work?

Yes, research supports the potential utility of the proposed NSD, and this section highlights some of the major findings from these studies, although this review is not intended to be exhaustive.

First, considerable evidence exists for a biological basis for the risk of death due to suicide. The concordance rates for suicide are 10 times higher in monozygotic (“identical”) vs dizygotic (“fraternal”) twins (24.1% vs 2.8%) and 2 to 5 times higher in relatives of those who die by suicide than in the general population. Heritability estimates of fatal suicides and nonfatal suicide attempts in biological relatives of adoptees who die from suicide range from 17% to 45%.11

Second, studies using information from small samples that was arduously collected by individual research groups have yielded important positive data. Most recently, in 2015, a multidisciplinary group led by Niculescu et al5 at Indiana University and other institutions described a test that could predict suicidality in men. This test was developed on the basis of a within-participant discovery approach to identify genes that change in expression between states of no suicidal ideation and high suicidal ideation, which was combined with clinical information assessed by 2 scales, the Convergent Functional Information for Suicidality and the Simplified Affective State Scale. Gene expression was measured in whole blood collected postmortem unless the method of suicide involved a medication overdose that could affect gene expression. These researchers identified 76 genes that likely were involved in suicidal ideation and suicide.

This report had a number of limitations.5 All of the individuals in these studies were being treated for psychiatric illness, were being closely followed by the investigators, and all were male. In addition, as noted above, suicides by overdose were eliminated from the analysis.

In a subsequent study published in 2016, the Niculescu group6 extended their work to women and identified 50 genes contributing to suicide risk in women. Underscoring the need for larger samples, only 3 of the top contributing genes were seen in both men and women, suggesting that there are likely significant sex differences in the biology of suicide completion. This important work needs to be replicated and extended.

In addition to these remarkable advances made in genetic understanding of the risk of suicide, recent research also has demonstrated a role for epigenetic and inflammatory processes as contributors to suicide risk.12-15

There are likely many contributors, including genetic, epigenetic, and environmental factors such as inflammatory processes, that increase the risk of suicide. The goal of this article is not to provide an exhaustive or integrative review of research in this area but rather to argue for the establishment of a national initiative to study all of these factors and to begin that process by establishing the NSD.

 

 

What will be the foreseeable outcome of this initiative?

The establishment of the NSD is expected to lead to better identification of those who are genetically at increased risk of suicide as well as biological factors (eg, inflammatory or other processes) and environmental factors (eg, drug abuse), which can turn that genetic risk into reality. Using research results made possible by the implementation of this proposal, objective testing can be developed to monitor risk more effectively than is currently possible using clinical assessment alone.

Furthermore, this work also can provide targets for developing new treatments. For example, there is convergence between the work of Niculescu et al,5,6 who identified genetic biomarkers for mechanistic target of rapamycin (mTOR) signaling as a risk factor in individuals who died by suicide and the work of Li et al and other researchers,16-18 whose findings have implicated mTOR-dependent synapse formation as a mechanism underlying the rapid (ie, within hours to a couple of days) antidepressant effects of N-methyl-d-aspartate antagonists, such as ketamine, CP-101,606, and esketamine. In fact, the authors of a study presented earlier this year reported that esketamine—an active enantiomer of ketamine—rapidly reduced suicidal ideation as well as other depressive symptoms in individuals admitted to the hospital for suicidal ideation.19 (mTOR is a serine/threonine protein kinase that regulates a number of biological processes in addition to synaptogenesis, including cell growth, cell proliferation, cell motility, cell survival, protein synthesis, and autophagy.20,21)

In aggregate, establishment of this proposed database will facilitate identification of biological (and therefore pharmaceutical) mechanisms beyond those involving biogenic amines, which have been the exclusive biological targets for antidepressants for the past 50 years.22 The likely consequences of the findings generated from research made possible by the proposed NSD will open completely new vistas for helping people at risk for suicide and psychiatric illnesses.

What foreseeable obstacles will need to be addressed?

Of course, obstacles and problems will arise but these will not exceed those encountered by the War on Cancer and they can similarly be overcome with sufficient public support and cooperation. Potential obstacles include:

  • need for incremental funding
  • obtaining the cooperation of the offices of each county medical examiner or coroner in a process that includes uniform systematic data collection
  • determining the situations (eg, time after death and means of death) that will allow for meaningful collection of data such as RNA and inflammatory biomarkers
  • establishing how data and particularly biological samples will be transported and stored
  • issues related to privacy of health information particularly for relatives of suicide victims
  • ensuring the reliability, validity, and comparability of the data received from different medical examiners and coroners.

With regard to the last issue, because stigma is associated with death by suicide, some true suicides could be missed, which would compromise sensitivity but simultaneously increase specificity. Other obstacles or problems may arise; however, I am certain that all such issues are surmountable and that the resulting NSD will be much better than what we have now and will propel our understanding of the biological underpinnings of the loss of life to suicide. (The author proposed a similar but even more ambitious plan 25 years ago,23 but he believes that this is an idea whose time has come.)

Acknowledgments

The author thanks Wayne C. Drevets, MD, Alexander Niculescu, MD, PhD, John Oldman, MD, and John Savitz, PhD, David Sheehan, MD, and Matthew Macaluso, DO for their review and suggestions concerning this proposal/manuscript, and Kaylee Hervey, MPH, from the Sedgwick County Health Department, Wichita, Kansas, for her input. The author also thanks Ruth Ross, as always, for her excellent editing and general assistance.

References

1. Pompili M, Gonda X, Serafini G, et al. Epidemiology of suicide in bipolar disorders: a systematic review of the literature. Bipolar Disord. 2013;15(5):457-490.
2. National Vital Statistics System; National Center for Health Statistics; Centers for Disease Control and Prevention. Ten leading causes of death by age group, United States–2014. Centers for Disease Control and Prevention. http://www.cdc.gov/injury/images/lc-charts/leading_causes_of_death_age_group_2014_1050w760h.gif. Accessed October 17, 2016.
3. Curtin SC, Warner M, Hedegaard H, et al. Increase in suicide in the United States, 1999-2014. National Center for Health Statistics Data Brief No. 241. Atlanta GA: National Center for Health Statistics, U.S. Department of Health and Human Services. http://www.cdc.gov/nchs/products/databriefs/db241.htm. Published April 2016. Accessed June 30, 2016.
4. Committee for the Workshop on the Medicolegal Death Investigation System; Board on Health Promotion and Disease Prevention. Medicolegal death investigation system: workshop summary. Washington, DC: National Academies Press; 2003.
5. Niculescu AB, Levey DF, Phalen PL, et al. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol Psychiatry. 2015;20(11):1266-1285.
6. Levey DF, Niculescu EM, Le-Niculescu H, et al. Towards understanding and predicting suicidality in women: biomarkers and clinical risk assessment. Mol Psychiatry. 2016;21(6):768-785.
7. World Health Organization. Prevention of suicide: guidelines for the formulation and implementation of national strategies. Geneva, Switzerland: World Health Organization; 1996.
8. U.S. Public Health Service. The Surgeon General’s call to action to prevent suicide. Washington, DC: U.S. Public Health Service; 1999.
9. U.S. Department of Health and Human Services (HHS). National Strategy for Suicide Prevention: goals and objectives for action. Rockville, MD: U.S. Department of Health and Human Services; 2001.
10. U.S. Department of Health and Human Services (HHS). National Strategy for Suicide Prevention: goals and objectives for action. Rockville, MD; U.S. Department of Health and Human Services; 2012.
11. Brent DA, Melham N. Familial transmission of suicidal behavior. Psychiatr Clin North Am. 2008;31(2):157-177.
12. Guintivano J, Brown T, Newcomer A, et al. Identification and replication of a combined epigenetic and genetic biomarker predicting suicide and suicidal behaviors. Am J Psychiatry. 2014;171(12):1287-1296.
13. Bay-Richter C, Linderholm KR, Lim CK, et al. A role for inflammatory metabolites as modulators of the glutamate N-methyl-D-aspartate receptor in depression and suicidality. Brain Behav Immun. 2015;43:110-117.
14. Brundin L, Bryleva EY, Thirtamara Rajamani K. Role of inflammation in suicide: from mechanisms to treatment [published online July 27, 2016]. Neuropsychopharmacology. doi: 10.1038/npp.2016.116.
15. Steiner J, Walter M, Gos T, et al. Severe depression is associated with increased microglial quinolinic acid in subregions of the anterior cingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission? J Neuroinflammation. 2011;8:94.
16. Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329(5994):959-964.
17. Zarate CA Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856-864.
18. Preskorn SH, Baker B, Kolluri S, et al. An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-D-aspartate antagonist, CP-101,606, in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol. 2008;28(6):631-637.
19. Canuso C, Singh J, Fedgchin M, et al. PeRSEVERe: a study of esketamine for the rapid reduction of the symptoms of major depressive disorder, including suicidal ideation, in subjects assessed to be at imminent risk for suicide. Presentation at the Annual Meeting of the American Society of Clinical Psychopharmacology, Scottsdale AZ, May 30-June 3, 2016.
20. Brown EJ, Albers MW, Shin TB, et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature. 1994;369(6483):756-758.
21. Moore PA, Rosen CA, Carter KC. Assignment of the human FKBP12-rapamycin-associated protein (FRAP) gene to chromosome 1p36 by fluorescence in situ hybridization. Genomics. 1996;33(2):331-332.
22. Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev. 2004;18(16):1926-1945.
23. Preskorn SH. The future and psychopharmacology: potentials and needs. Psychiatr Ann. 1990;20(11):625-633.

References

1. Pompili M, Gonda X, Serafini G, et al. Epidemiology of suicide in bipolar disorders: a systematic review of the literature. Bipolar Disord. 2013;15(5):457-490.
2. National Vital Statistics System; National Center for Health Statistics; Centers for Disease Control and Prevention. Ten leading causes of death by age group, United States–2014. Centers for Disease Control and Prevention. http://www.cdc.gov/injury/images/lc-charts/leading_causes_of_death_age_group_2014_1050w760h.gif. Accessed October 17, 2016.
3. Curtin SC, Warner M, Hedegaard H, et al. Increase in suicide in the United States, 1999-2014. National Center for Health Statistics Data Brief No. 241. Atlanta GA: National Center for Health Statistics, U.S. Department of Health and Human Services. http://www.cdc.gov/nchs/products/databriefs/db241.htm. Published April 2016. Accessed June 30, 2016.
4. Committee for the Workshop on the Medicolegal Death Investigation System; Board on Health Promotion and Disease Prevention. Medicolegal death investigation system: workshop summary. Washington, DC: National Academies Press; 2003.
5. Niculescu AB, Levey DF, Phalen PL, et al. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol Psychiatry. 2015;20(11):1266-1285.
6. Levey DF, Niculescu EM, Le-Niculescu H, et al. Towards understanding and predicting suicidality in women: biomarkers and clinical risk assessment. Mol Psychiatry. 2016;21(6):768-785.
7. World Health Organization. Prevention of suicide: guidelines for the formulation and implementation of national strategies. Geneva, Switzerland: World Health Organization; 1996.
8. U.S. Public Health Service. The Surgeon General’s call to action to prevent suicide. Washington, DC: U.S. Public Health Service; 1999.
9. U.S. Department of Health and Human Services (HHS). National Strategy for Suicide Prevention: goals and objectives for action. Rockville, MD: U.S. Department of Health and Human Services; 2001.
10. U.S. Department of Health and Human Services (HHS). National Strategy for Suicide Prevention: goals and objectives for action. Rockville, MD; U.S. Department of Health and Human Services; 2012.
11. Brent DA, Melham N. Familial transmission of suicidal behavior. Psychiatr Clin North Am. 2008;31(2):157-177.
12. Guintivano J, Brown T, Newcomer A, et al. Identification and replication of a combined epigenetic and genetic biomarker predicting suicide and suicidal behaviors. Am J Psychiatry. 2014;171(12):1287-1296.
13. Bay-Richter C, Linderholm KR, Lim CK, et al. A role for inflammatory metabolites as modulators of the glutamate N-methyl-D-aspartate receptor in depression and suicidality. Brain Behav Immun. 2015;43:110-117.
14. Brundin L, Bryleva EY, Thirtamara Rajamani K. Role of inflammation in suicide: from mechanisms to treatment [published online July 27, 2016]. Neuropsychopharmacology. doi: 10.1038/npp.2016.116.
15. Steiner J, Walter M, Gos T, et al. Severe depression is associated with increased microglial quinolinic acid in subregions of the anterior cingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission? J Neuroinflammation. 2011;8:94.
16. Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329(5994):959-964.
17. Zarate CA Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856-864.
18. Preskorn SH, Baker B, Kolluri S, et al. An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-D-aspartate antagonist, CP-101,606, in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol. 2008;28(6):631-637.
19. Canuso C, Singh J, Fedgchin M, et al. PeRSEVERe: a study of esketamine for the rapid reduction of the symptoms of major depressive disorder, including suicidal ideation, in subjects assessed to be at imminent risk for suicide. Presentation at the Annual Meeting of the American Society of Clinical Psychopharmacology, Scottsdale AZ, May 30-June 3, 2016.
20. Brown EJ, Albers MW, Shin TB, et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature. 1994;369(6483):756-758.
21. Moore PA, Rosen CA, Carter KC. Assignment of the human FKBP12-rapamycin-associated protein (FRAP) gene to chromosome 1p36 by fluorescence in situ hybridization. Genomics. 1996;33(2):331-332.
22. Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev. 2004;18(16):1926-1945.
23. Preskorn SH. The future and psychopharmacology: potentials and needs. Psychiatr Ann. 1990;20(11):625-633.

Issue
November 2016
Issue
November 2016
Page Number
24,30-32,36-37
Page Number
24,30-32,36-37
Publications
Publications
Topics
Article Type
Display Headline
National Initiative to Prevent Suicide: A new proposal to improve the understanding and prevention of suicide
Display Headline
National Initiative to Prevent Suicide: A new proposal to improve the understanding and prevention of suicide
Sections
Disallow All Ads
Article PDF Media

Genetic and related laboratory tests in psychiatry: What mental health practitioners need to know

Article Type
Changed
Thu, 03/28/2019 - 15:10
Display Headline
Genetic and related laboratory tests in psychiatry: What mental health practitioners need to know

What has been the history of the development of laboratory tests in the field of psychiatry?

During my almost-40-year academic medical career, I have been interested in the development and incorporation of laboratory tests into psychiatry.1 This interest initially focused on therapeutic drug monitoring (TDM) and the genetics of drug responsiveness, with an emphasis on drug metabolism. In addition to TDM—which I have long believed is vastly underutilized in psychiatry—there have been many failed attempts to develop diagnostic tests, including tests to distinguish between what were postulated to be serotonergic and noradrenergic forms of major depression in the 1970s2,3 and the dexamethasone suppression test for melancholia in the 1980s.4 Recently, a 51-analyte immunoassay test was marketed by Rules-Based Medicine, Inc. (RBM), as an aid in the diagnosis of schizophrenia, but the test was found to suffer a high false-positive rate and was withdrawn from the market.5 Given this track record, caution is warranted when examining claims for new tests.


What types of tests are being developed?
Most tests in development are pharmacogenomic (PG)-based or immunoassay (IA)-based.

PG tests examine single nucleotide polymorphisms (SNP) in genes that code for pharmacokinetic mechanisms, primarily cytochrome P450 (CYP) enzymes responsible for drug metabolism and P-glycoprotein, responsible for drug transportation. The next most common type of test examines pharmacodynamic mechanisms, such as SNPs of specific receptor genes, including serotonin (or 5-hydroxytryptophan [5-HT] transporter [SET or 5-HTT]) or the 5-HT2A receptor.

The fact that CYP enzymes lead the list is not surprising: These enzymes and their role in the metabolism of specific drugs have been extensively studied since the late 1980s. Considerable data has been accumulated regarding variants of CYP enzymes, which convey clinically meaningful differences among individuals in terms of their ability to metabolize drug via these pathways. Individuals are commonly divided into 4 phenotypic categories: ultra-rapid, extensive (or normal), intermediate, and poor metabolizers. Based on these phenotypes, clinical consequences can be quantitated in terms of changes in drug concentration, concentration-dependent beneficial or adverse effects, and associated/recommended changes in dosing.

Research into the role of pharmacodynamic variants, however, is still in infancy and more difficult to measure in terms of assessing endpoints, with related limitations in clinical utility.

IA assays generally measure a variety of proteins, particularly those reflecting inflammatory processes (eg, various cytokines, such as interleukin-6).6 As with pharmacodynamic measures, research into the role of inflammatory biomarkers is in early stages. The clinical utility of associated tests is, therefore, less certain; witness the recent study5 I noted that revealed a high false-positive rate for the RBM schizophrenia panel in healthy controls. Nevertheless, considerable research is being conducted in all of these areas so that new developments might lend themselves to greater clinical utility.

(Note that PG biomarkers are trait measures, whereas IA biomarkers are state measures, so that complementary use of both types of tests might prove useful in diagnosis and clinical management. Although such integrative use of these 2 different types of tests generally is not done today.)


What does it take to market these tests?
At a minimum, offering these tests for sale requires that the laboratory be certified by the Centers for Medicare & Medicaid Services, according to the Clinical Laboratory Improvement Amendments (CLIA) standards (www.fda.gov/medicaldevices/deviceregulationandguidance/ivdregulatoryassistance/ucm124105.htm). CLIA-certified laboratories are required to demonstrate the analytical validity of tests that they offer—ie, the accuracy and reliability of the test in measuring a parameter of interest—but not the clinical validity or utility of those tests. The fact that a test in fact measures what it claims to be measuring in and of itself does not mean it has clinical validity or utility (see the discussion below).


Must the FDA approve laboratory tests?
No, but that situation might be changing.

Currently, only tests used in a setting considered high risk—eg, a test intended to detect or diagnose a malignancy or guide its treatment—requires formal FDA approval. The approval of such a test requires submission to the FDA of clinical data supporting its clinical validity and utility, in addition to evidence of analytic validity.

Even in such cases, the degree and quality of the clinical data required are generally not as high as would be required for approval of a drug. That distinction is understandable, given the type and quantity of data necessary for drug approval and the many years and billions of dollars it takes to accumulate such data. For most laboratory tests, providing the same level of data required to have a drug approved would be neither necessary nor feasible given the business model underlying most laboratories providing laboratory tests.

 

 


What do ‘clinical validity’ and ‘clinical utility’ mean?
These are higher evidence thresholds than is needed for analytic validity, although the latter is a necessary first step on the path to achieving these higher thresholds.

Clinical validity is the ability of a test to detect:

 

  • a clinically meaningful measure, such as clinical response
  • an adverse effect
  • a biologically meaningful measure (eg, a drug level or a change in the electrocardiographic pattern).

Above the threshold of clinical validity is clinical utility, which is proof that the test can reliably be used to guide clinical management and thus meaningfully improve outcomes, such as guiding drug or dosage selection.


Is the use of PG testing recommended? If so, in what instances?
Specific types of PG testing is recommended by the FDA recommended. The FDA has been incorporating PG information into the labels of specific medications for several years; the agency has a Web site (www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/ucm083378.htm) that continuously updates this information. The involved drugs are in all therapeutic classes—from oncology to psychiatry.

More than 30 psychotropic drugs have PG information in their label; some of those drugs’ labels contain specific recommendations, such as obtaining PG information before selecting or starting a drug in a specific patient. An example is carbamazepine, for which the recommendation is to obtain HLA testing before starting the drug in patients of Han Chinese ancestry, because members of this large ethnic group are at greater risk of serious dermatologic adverse effects, including Stevens-Johnson syndrome.

In other instances, the recommendation is to do the testing before increasing beyond a specific dose. Examples of psychiatric drugs whose labels contain such PG information include pimozide and iloperidone as well as citalopram. In the FDA-approved label, guidance is provided that these drugs can be started without testing if prescribed at a reduced recommended starting dosage range, rather than the full starting dosage range. The guidance on these drugs further recommends testing for genetic CYP2D6 poor metabolizer (PM) status before dosing above that initial recommended, limited, starting dosage range.

The rationale for this guidance is to reduce the risk that (1) patients in question will achieve an excessively high plasma drug level that can cause significant prolongation of intracardiac conduction (eg, QTc prolongation) and thus (2) develop the potentially fatal arrhythmia torsades de pointes. Guidance is based on thorough QTc studies that were performed on each drug,7,8 which makes them examples of instances in which the test has clinical validity and utility as well as analytical validity.

To find PG labeling in the package insert for these drugs, visit: www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm.


What about data for other tests that are marketed and promoted by developers?
Sometimes, there are—literally—no data on available tests beyond the analytical validity of the test; other times, the amount and quality of clinical data are quite variable, ranging from results of ≥1 small retrospective studies without controls to results of prospective, randomized, controlled studies. Even among the latter, the developer may conduct and analyze their studies without oversight by an independent agency, such as the FDA.

This situation (1) raises concern that study results are not independent of the developer’s business interests and, as one might expect, (2) leads to controversy about whether the data are compelling—or not.9-12


What is a critical difference between PG test results and results of most laboratory tests?
PG tests are, as noted, trait rather than state characteristics. That means that the results do not change except for a phenomenon known as phenocoversion, discussed below. (Of course, advances in gene therapy might make it possible someday to change a person’s genetic makeup and for mitochondrial genes that is already possible.) 

For this reason, PG test results should not get buried in the medical record, as might happen with, say, a patient’s serum potassium level at a given point in time. Instead, PG test results need to be carried forward continuously. Results also should be given to the patient as part of his (her) personal health record and to all other health care providers that the patient is seeing or will see in the future. Each health care provider who obtains PG test results should consider sending them to all current clinicians providing care for the patient at the same time as they are.


Is your functional status at a given moment the same as your genetic status?
No. There is a phenomenon known as phenoconversion in which a person’s current functional status may be different from what would be expected based on their genetic status.

CYP2D6 functional status is susceptible to phenoconversion as follows: Administering fluoxetine and paroxetine, for example, at 20 or 40 mg/d converts 66% and 95%, respectively, of patients who are CYP2D6 extensive (ie, normal) metabolizers into phenocopies of people who, genetically, lack the ability to metabolize drugs via CYP2D6 (ie, genotypic CYP2D6 PM). Based on a recent study of 900 participants in routine clinical care who were taking an antidepressant, 4% of the general U.S. population are genetically CYP2D6 PM; an additional 24% are phenotypically CYP2D6 PM because of concomitant administration of a CYP2D6 substantial inhibitor, such as bupropion, fluoxetine, paroxetine, or terbenafine.13

 

 

That is the reason a provider needs to know what drugs a patient is taking concomitantly—to consider the possibility of phenoconversion and, when necessary, to dose accordingly.


What does the future hold?
Development of tests for use in psychiatric practice is likely to grow substantially, for at least 2 reasons:

 

  • There is a huge unmet need for clinically meaningful tests to aid in the provision of optimal patient care and, therefore, a tremendous business opportunity
  • Knowledge in the biological basis of psychiatric disorders is growing exponentially; with that knowledge comes the ability to develop new tests.

A recent example comes from a research group that devised a test that could predict suicidality.14 Time will tell whether this test or a derivative of it enters practice. Nevertheless, it is a harbinger of the likely dramatic changes in the landscape of clinical medicine particularly as it applies to psychiatry.

Given these developments, the syndromic diagnoses in DSM-5 will in the future likely be replaced by a new diagnostic schema that breaks down existing heterogenous syndromic diagnoses into pathophysiologically and etiologically meaningful entities using insights gained from genetic and biomarker data as well as functional brain imaging. Theoretically, those insights will lead to new modalities of treatment, including somatic treatments that target novel mechanisms of action, coupled to more effective psychosocial therapies—with both therapies guided by diagnostic tests to monitor response to specific treatment interventions.

During this transition from the past to the future, answers to the questions I’ve posed here about laboratory testing in psychiatry will, I hope, help the practitioner understand, evaluate, and incorporate these changes readily into practice.

 


Disclosure
The author serves on the scientific advisory board of Assurex Health, the developer and marketer of the GeneSight test.
References


1. Preskorn SH, Biggs JT. Use of tricyclic antidepressant blood levels. N Engl J Med. 1978;298(3):166.
2. Schildkraut JJ. Biogenic amines and affective disorders. Annu Rev Med. 1974;25(0):333-348.
3. Maas JW. Biogenic amines and depression. Biochemical and pharmacological separation of two types of depression. Arch Gen Psychiatry. 1975;32(11):1357-1361.
4. Carroll BJ, Feinberg M, Greden JF, et al. A specific laboratory test for the diagnosis of melancholia. Standardization, validation, and clinical utility. Arch Gen Psychiatry. 1981;38(1):15-22.
5. Wehler C, Preskorn S. High false-positive rate of a putative biomarker test to aid in the diagnosis of schizophrenia. J Clin Psychiatry. In press.
6. Savitz J, Preskorn S, Teague TK, et al. Minocycline and aspirin in the treatment of bipolar depression: a protocol for a proof-of-concept, randomised, double-blind, placebo-controlled, 2x2 clinical trial. BMJ Open. 2012;2(1):e000643. doi: 10.1136/bmjopen-2011-000643.
7. Rogers HL, Bhattaram A, Zineh I, et al. CYP2D6 genotype information to guide pimozide treatment in adult and pediatric patients: basis for the U.S. Food and Drug Administration’s new dosing recommendations. J Clin Psychiatry. 2012;73(9):1187-1190.
8. Potkin S, Preskorn S, Hochfeld M, et al. A thorough QTc study of 3 doses of iloperidone including metabolic inhibition via CYP2D6 and/or CYP3A4 inhibition and a comparison to quetiapine and ziprasidone. J Clin Psychopharmacol. 2013;33(1):3-10.
9. Howland RH. Pharmacogenetic testing in psychiatry: not (quite) ready for primetime. J Psychosoc Nurs Ment Health Serv. 2014;52(11):13-16.
10. Rosenblat JD, Lee Y, McIntyre RS. Does pharmacogenomics testing improve clinical outcomes for major depressive disorder? A systematic review of clinical trials and cost-effectiveness studies. J Clin Psychiatry. In press.
11. Nassan M, Nicholson WT, Elliott MA, et al. Pharmacokinetic pharmacogenetic prescribing guidelines for antidepressants: a template for psychiatric precision medicine. Mayo Clin Proc. In press.
12. Altar CA, Carhart JM, Allen JD, et al. Clinical validity: combinatorial pharmacogenomics predicts antidepressant responses and healthcare utilizations better than single gene phenotypes. Pharmacogenomics J. 2015;15(5):443-451.
13. Preskorn S, Kane C, Lobello K, et al. Cytochrome P450 2D6 phenoconversion is common in patients being treated for depression: implications for personalized medicine. J Clin Psychiatry. 2013;74(6):614-621.
14. Niculescu AB, Levey DF, Phalen PL, et al. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol Psychiatry. 2015;20(11):1266-1285.

Article PDF
Author and Disclosure Information

 

Sheldon H. Preskorn, MD
Professor of Psychiatry
University of Kansas School of Medicine-Wichita
Wichita, Kansas
President and Chief Executive Officer
World-Wide Psychopharmacology Consultants
He is a member of the Current Psychiatry Editorial Board

Issue
Current Psychiatry - 15(4)
Publications
Topics
Page Number
19-22,58
Legacy Keywords
genetics, therapeutic drug monitoring, pharmacogenomic, immunoassay
Sections
Author and Disclosure Information

 

Sheldon H. Preskorn, MD
Professor of Psychiatry
University of Kansas School of Medicine-Wichita
Wichita, Kansas
President and Chief Executive Officer
World-Wide Psychopharmacology Consultants
He is a member of the Current Psychiatry Editorial Board

Author and Disclosure Information

 

Sheldon H. Preskorn, MD
Professor of Psychiatry
University of Kansas School of Medicine-Wichita
Wichita, Kansas
President and Chief Executive Officer
World-Wide Psychopharmacology Consultants
He is a member of the Current Psychiatry Editorial Board

Article PDF
Article PDF

What has been the history of the development of laboratory tests in the field of psychiatry?

During my almost-40-year academic medical career, I have been interested in the development and incorporation of laboratory tests into psychiatry.1 This interest initially focused on therapeutic drug monitoring (TDM) and the genetics of drug responsiveness, with an emphasis on drug metabolism. In addition to TDM—which I have long believed is vastly underutilized in psychiatry—there have been many failed attempts to develop diagnostic tests, including tests to distinguish between what were postulated to be serotonergic and noradrenergic forms of major depression in the 1970s2,3 and the dexamethasone suppression test for melancholia in the 1980s.4 Recently, a 51-analyte immunoassay test was marketed by Rules-Based Medicine, Inc. (RBM), as an aid in the diagnosis of schizophrenia, but the test was found to suffer a high false-positive rate and was withdrawn from the market.5 Given this track record, caution is warranted when examining claims for new tests.


What types of tests are being developed?
Most tests in development are pharmacogenomic (PG)-based or immunoassay (IA)-based.

PG tests examine single nucleotide polymorphisms (SNP) in genes that code for pharmacokinetic mechanisms, primarily cytochrome P450 (CYP) enzymes responsible for drug metabolism and P-glycoprotein, responsible for drug transportation. The next most common type of test examines pharmacodynamic mechanisms, such as SNPs of specific receptor genes, including serotonin (or 5-hydroxytryptophan [5-HT] transporter [SET or 5-HTT]) or the 5-HT2A receptor.

The fact that CYP enzymes lead the list is not surprising: These enzymes and their role in the metabolism of specific drugs have been extensively studied since the late 1980s. Considerable data has been accumulated regarding variants of CYP enzymes, which convey clinically meaningful differences among individuals in terms of their ability to metabolize drug via these pathways. Individuals are commonly divided into 4 phenotypic categories: ultra-rapid, extensive (or normal), intermediate, and poor metabolizers. Based on these phenotypes, clinical consequences can be quantitated in terms of changes in drug concentration, concentration-dependent beneficial or adverse effects, and associated/recommended changes in dosing.

Research into the role of pharmacodynamic variants, however, is still in infancy and more difficult to measure in terms of assessing endpoints, with related limitations in clinical utility.

IA assays generally measure a variety of proteins, particularly those reflecting inflammatory processes (eg, various cytokines, such as interleukin-6).6 As with pharmacodynamic measures, research into the role of inflammatory biomarkers is in early stages. The clinical utility of associated tests is, therefore, less certain; witness the recent study5 I noted that revealed a high false-positive rate for the RBM schizophrenia panel in healthy controls. Nevertheless, considerable research is being conducted in all of these areas so that new developments might lend themselves to greater clinical utility.

(Note that PG biomarkers are trait measures, whereas IA biomarkers are state measures, so that complementary use of both types of tests might prove useful in diagnosis and clinical management. Although such integrative use of these 2 different types of tests generally is not done today.)


What does it take to market these tests?
At a minimum, offering these tests for sale requires that the laboratory be certified by the Centers for Medicare & Medicaid Services, according to the Clinical Laboratory Improvement Amendments (CLIA) standards (www.fda.gov/medicaldevices/deviceregulationandguidance/ivdregulatoryassistance/ucm124105.htm). CLIA-certified laboratories are required to demonstrate the analytical validity of tests that they offer—ie, the accuracy and reliability of the test in measuring a parameter of interest—but not the clinical validity or utility of those tests. The fact that a test in fact measures what it claims to be measuring in and of itself does not mean it has clinical validity or utility (see the discussion below).


Must the FDA approve laboratory tests?
No, but that situation might be changing.

Currently, only tests used in a setting considered high risk—eg, a test intended to detect or diagnose a malignancy or guide its treatment—requires formal FDA approval. The approval of such a test requires submission to the FDA of clinical data supporting its clinical validity and utility, in addition to evidence of analytic validity.

Even in such cases, the degree and quality of the clinical data required are generally not as high as would be required for approval of a drug. That distinction is understandable, given the type and quantity of data necessary for drug approval and the many years and billions of dollars it takes to accumulate such data. For most laboratory tests, providing the same level of data required to have a drug approved would be neither necessary nor feasible given the business model underlying most laboratories providing laboratory tests.

 

 


What do ‘clinical validity’ and ‘clinical utility’ mean?
These are higher evidence thresholds than is needed for analytic validity, although the latter is a necessary first step on the path to achieving these higher thresholds.

Clinical validity is the ability of a test to detect:

 

  • a clinically meaningful measure, such as clinical response
  • an adverse effect
  • a biologically meaningful measure (eg, a drug level or a change in the electrocardiographic pattern).

Above the threshold of clinical validity is clinical utility, which is proof that the test can reliably be used to guide clinical management and thus meaningfully improve outcomes, such as guiding drug or dosage selection.


Is the use of PG testing recommended? If so, in what instances?
Specific types of PG testing is recommended by the FDA recommended. The FDA has been incorporating PG information into the labels of specific medications for several years; the agency has a Web site (www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/ucm083378.htm) that continuously updates this information. The involved drugs are in all therapeutic classes—from oncology to psychiatry.

More than 30 psychotropic drugs have PG information in their label; some of those drugs’ labels contain specific recommendations, such as obtaining PG information before selecting or starting a drug in a specific patient. An example is carbamazepine, for which the recommendation is to obtain HLA testing before starting the drug in patients of Han Chinese ancestry, because members of this large ethnic group are at greater risk of serious dermatologic adverse effects, including Stevens-Johnson syndrome.

In other instances, the recommendation is to do the testing before increasing beyond a specific dose. Examples of psychiatric drugs whose labels contain such PG information include pimozide and iloperidone as well as citalopram. In the FDA-approved label, guidance is provided that these drugs can be started without testing if prescribed at a reduced recommended starting dosage range, rather than the full starting dosage range. The guidance on these drugs further recommends testing for genetic CYP2D6 poor metabolizer (PM) status before dosing above that initial recommended, limited, starting dosage range.

The rationale for this guidance is to reduce the risk that (1) patients in question will achieve an excessively high plasma drug level that can cause significant prolongation of intracardiac conduction (eg, QTc prolongation) and thus (2) develop the potentially fatal arrhythmia torsades de pointes. Guidance is based on thorough QTc studies that were performed on each drug,7,8 which makes them examples of instances in which the test has clinical validity and utility as well as analytical validity.

To find PG labeling in the package insert for these drugs, visit: www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm.


What about data for other tests that are marketed and promoted by developers?
Sometimes, there are—literally—no data on available tests beyond the analytical validity of the test; other times, the amount and quality of clinical data are quite variable, ranging from results of ≥1 small retrospective studies without controls to results of prospective, randomized, controlled studies. Even among the latter, the developer may conduct and analyze their studies without oversight by an independent agency, such as the FDA.

This situation (1) raises concern that study results are not independent of the developer’s business interests and, as one might expect, (2) leads to controversy about whether the data are compelling—or not.9-12


What is a critical difference between PG test results and results of most laboratory tests?
PG tests are, as noted, trait rather than state characteristics. That means that the results do not change except for a phenomenon known as phenocoversion, discussed below. (Of course, advances in gene therapy might make it possible someday to change a person’s genetic makeup and for mitochondrial genes that is already possible.) 

For this reason, PG test results should not get buried in the medical record, as might happen with, say, a patient’s serum potassium level at a given point in time. Instead, PG test results need to be carried forward continuously. Results also should be given to the patient as part of his (her) personal health record and to all other health care providers that the patient is seeing or will see in the future. Each health care provider who obtains PG test results should consider sending them to all current clinicians providing care for the patient at the same time as they are.


Is your functional status at a given moment the same as your genetic status?
No. There is a phenomenon known as phenoconversion in which a person’s current functional status may be different from what would be expected based on their genetic status.

CYP2D6 functional status is susceptible to phenoconversion as follows: Administering fluoxetine and paroxetine, for example, at 20 or 40 mg/d converts 66% and 95%, respectively, of patients who are CYP2D6 extensive (ie, normal) metabolizers into phenocopies of people who, genetically, lack the ability to metabolize drugs via CYP2D6 (ie, genotypic CYP2D6 PM). Based on a recent study of 900 participants in routine clinical care who were taking an antidepressant, 4% of the general U.S. population are genetically CYP2D6 PM; an additional 24% are phenotypically CYP2D6 PM because of concomitant administration of a CYP2D6 substantial inhibitor, such as bupropion, fluoxetine, paroxetine, or terbenafine.13

 

 

That is the reason a provider needs to know what drugs a patient is taking concomitantly—to consider the possibility of phenoconversion and, when necessary, to dose accordingly.


What does the future hold?
Development of tests for use in psychiatric practice is likely to grow substantially, for at least 2 reasons:

 

  • There is a huge unmet need for clinically meaningful tests to aid in the provision of optimal patient care and, therefore, a tremendous business opportunity
  • Knowledge in the biological basis of psychiatric disorders is growing exponentially; with that knowledge comes the ability to develop new tests.

A recent example comes from a research group that devised a test that could predict suicidality.14 Time will tell whether this test or a derivative of it enters practice. Nevertheless, it is a harbinger of the likely dramatic changes in the landscape of clinical medicine particularly as it applies to psychiatry.

Given these developments, the syndromic diagnoses in DSM-5 will in the future likely be replaced by a new diagnostic schema that breaks down existing heterogenous syndromic diagnoses into pathophysiologically and etiologically meaningful entities using insights gained from genetic and biomarker data as well as functional brain imaging. Theoretically, those insights will lead to new modalities of treatment, including somatic treatments that target novel mechanisms of action, coupled to more effective psychosocial therapies—with both therapies guided by diagnostic tests to monitor response to specific treatment interventions.

During this transition from the past to the future, answers to the questions I’ve posed here about laboratory testing in psychiatry will, I hope, help the practitioner understand, evaluate, and incorporate these changes readily into practice.

 


Disclosure
The author serves on the scientific advisory board of Assurex Health, the developer and marketer of the GeneSight test.

What has been the history of the development of laboratory tests in the field of psychiatry?

During my almost-40-year academic medical career, I have been interested in the development and incorporation of laboratory tests into psychiatry.1 This interest initially focused on therapeutic drug monitoring (TDM) and the genetics of drug responsiveness, with an emphasis on drug metabolism. In addition to TDM—which I have long believed is vastly underutilized in psychiatry—there have been many failed attempts to develop diagnostic tests, including tests to distinguish between what were postulated to be serotonergic and noradrenergic forms of major depression in the 1970s2,3 and the dexamethasone suppression test for melancholia in the 1980s.4 Recently, a 51-analyte immunoassay test was marketed by Rules-Based Medicine, Inc. (RBM), as an aid in the diagnosis of schizophrenia, but the test was found to suffer a high false-positive rate and was withdrawn from the market.5 Given this track record, caution is warranted when examining claims for new tests.


What types of tests are being developed?
Most tests in development are pharmacogenomic (PG)-based or immunoassay (IA)-based.

PG tests examine single nucleotide polymorphisms (SNP) in genes that code for pharmacokinetic mechanisms, primarily cytochrome P450 (CYP) enzymes responsible for drug metabolism and P-glycoprotein, responsible for drug transportation. The next most common type of test examines pharmacodynamic mechanisms, such as SNPs of specific receptor genes, including serotonin (or 5-hydroxytryptophan [5-HT] transporter [SET or 5-HTT]) or the 5-HT2A receptor.

The fact that CYP enzymes lead the list is not surprising: These enzymes and their role in the metabolism of specific drugs have been extensively studied since the late 1980s. Considerable data has been accumulated regarding variants of CYP enzymes, which convey clinically meaningful differences among individuals in terms of their ability to metabolize drug via these pathways. Individuals are commonly divided into 4 phenotypic categories: ultra-rapid, extensive (or normal), intermediate, and poor metabolizers. Based on these phenotypes, clinical consequences can be quantitated in terms of changes in drug concentration, concentration-dependent beneficial or adverse effects, and associated/recommended changes in dosing.

Research into the role of pharmacodynamic variants, however, is still in infancy and more difficult to measure in terms of assessing endpoints, with related limitations in clinical utility.

IA assays generally measure a variety of proteins, particularly those reflecting inflammatory processes (eg, various cytokines, such as interleukin-6).6 As with pharmacodynamic measures, research into the role of inflammatory biomarkers is in early stages. The clinical utility of associated tests is, therefore, less certain; witness the recent study5 I noted that revealed a high false-positive rate for the RBM schizophrenia panel in healthy controls. Nevertheless, considerable research is being conducted in all of these areas so that new developments might lend themselves to greater clinical utility.

(Note that PG biomarkers are trait measures, whereas IA biomarkers are state measures, so that complementary use of both types of tests might prove useful in diagnosis and clinical management. Although such integrative use of these 2 different types of tests generally is not done today.)


What does it take to market these tests?
At a minimum, offering these tests for sale requires that the laboratory be certified by the Centers for Medicare & Medicaid Services, according to the Clinical Laboratory Improvement Amendments (CLIA) standards (www.fda.gov/medicaldevices/deviceregulationandguidance/ivdregulatoryassistance/ucm124105.htm). CLIA-certified laboratories are required to demonstrate the analytical validity of tests that they offer—ie, the accuracy and reliability of the test in measuring a parameter of interest—but not the clinical validity or utility of those tests. The fact that a test in fact measures what it claims to be measuring in and of itself does not mean it has clinical validity or utility (see the discussion below).


Must the FDA approve laboratory tests?
No, but that situation might be changing.

Currently, only tests used in a setting considered high risk—eg, a test intended to detect or diagnose a malignancy or guide its treatment—requires formal FDA approval. The approval of such a test requires submission to the FDA of clinical data supporting its clinical validity and utility, in addition to evidence of analytic validity.

Even in such cases, the degree and quality of the clinical data required are generally not as high as would be required for approval of a drug. That distinction is understandable, given the type and quantity of data necessary for drug approval and the many years and billions of dollars it takes to accumulate such data. For most laboratory tests, providing the same level of data required to have a drug approved would be neither necessary nor feasible given the business model underlying most laboratories providing laboratory tests.

 

 


What do ‘clinical validity’ and ‘clinical utility’ mean?
These are higher evidence thresholds than is needed for analytic validity, although the latter is a necessary first step on the path to achieving these higher thresholds.

Clinical validity is the ability of a test to detect:

 

  • a clinically meaningful measure, such as clinical response
  • an adverse effect
  • a biologically meaningful measure (eg, a drug level or a change in the electrocardiographic pattern).

Above the threshold of clinical validity is clinical utility, which is proof that the test can reliably be used to guide clinical management and thus meaningfully improve outcomes, such as guiding drug or dosage selection.


Is the use of PG testing recommended? If so, in what instances?
Specific types of PG testing is recommended by the FDA recommended. The FDA has been incorporating PG information into the labels of specific medications for several years; the agency has a Web site (www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/ucm083378.htm) that continuously updates this information. The involved drugs are in all therapeutic classes—from oncology to psychiatry.

More than 30 psychotropic drugs have PG information in their label; some of those drugs’ labels contain specific recommendations, such as obtaining PG information before selecting or starting a drug in a specific patient. An example is carbamazepine, for which the recommendation is to obtain HLA testing before starting the drug in patients of Han Chinese ancestry, because members of this large ethnic group are at greater risk of serious dermatologic adverse effects, including Stevens-Johnson syndrome.

In other instances, the recommendation is to do the testing before increasing beyond a specific dose. Examples of psychiatric drugs whose labels contain such PG information include pimozide and iloperidone as well as citalopram. In the FDA-approved label, guidance is provided that these drugs can be started without testing if prescribed at a reduced recommended starting dosage range, rather than the full starting dosage range. The guidance on these drugs further recommends testing for genetic CYP2D6 poor metabolizer (PM) status before dosing above that initial recommended, limited, starting dosage range.

The rationale for this guidance is to reduce the risk that (1) patients in question will achieve an excessively high plasma drug level that can cause significant prolongation of intracardiac conduction (eg, QTc prolongation) and thus (2) develop the potentially fatal arrhythmia torsades de pointes. Guidance is based on thorough QTc studies that were performed on each drug,7,8 which makes them examples of instances in which the test has clinical validity and utility as well as analytical validity.

To find PG labeling in the package insert for these drugs, visit: www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm.


What about data for other tests that are marketed and promoted by developers?
Sometimes, there are—literally—no data on available tests beyond the analytical validity of the test; other times, the amount and quality of clinical data are quite variable, ranging from results of ≥1 small retrospective studies without controls to results of prospective, randomized, controlled studies. Even among the latter, the developer may conduct and analyze their studies without oversight by an independent agency, such as the FDA.

This situation (1) raises concern that study results are not independent of the developer’s business interests and, as one might expect, (2) leads to controversy about whether the data are compelling—or not.9-12


What is a critical difference between PG test results and results of most laboratory tests?
PG tests are, as noted, trait rather than state characteristics. That means that the results do not change except for a phenomenon known as phenocoversion, discussed below. (Of course, advances in gene therapy might make it possible someday to change a person’s genetic makeup and for mitochondrial genes that is already possible.) 

For this reason, PG test results should not get buried in the medical record, as might happen with, say, a patient’s serum potassium level at a given point in time. Instead, PG test results need to be carried forward continuously. Results also should be given to the patient as part of his (her) personal health record and to all other health care providers that the patient is seeing or will see in the future. Each health care provider who obtains PG test results should consider sending them to all current clinicians providing care for the patient at the same time as they are.


Is your functional status at a given moment the same as your genetic status?
No. There is a phenomenon known as phenoconversion in which a person’s current functional status may be different from what would be expected based on their genetic status.

CYP2D6 functional status is susceptible to phenoconversion as follows: Administering fluoxetine and paroxetine, for example, at 20 or 40 mg/d converts 66% and 95%, respectively, of patients who are CYP2D6 extensive (ie, normal) metabolizers into phenocopies of people who, genetically, lack the ability to metabolize drugs via CYP2D6 (ie, genotypic CYP2D6 PM). Based on a recent study of 900 participants in routine clinical care who were taking an antidepressant, 4% of the general U.S. population are genetically CYP2D6 PM; an additional 24% are phenotypically CYP2D6 PM because of concomitant administration of a CYP2D6 substantial inhibitor, such as bupropion, fluoxetine, paroxetine, or terbenafine.13

 

 

That is the reason a provider needs to know what drugs a patient is taking concomitantly—to consider the possibility of phenoconversion and, when necessary, to dose accordingly.


What does the future hold?
Development of tests for use in psychiatric practice is likely to grow substantially, for at least 2 reasons:

 

  • There is a huge unmet need for clinically meaningful tests to aid in the provision of optimal patient care and, therefore, a tremendous business opportunity
  • Knowledge in the biological basis of psychiatric disorders is growing exponentially; with that knowledge comes the ability to develop new tests.

A recent example comes from a research group that devised a test that could predict suicidality.14 Time will tell whether this test or a derivative of it enters practice. Nevertheless, it is a harbinger of the likely dramatic changes in the landscape of clinical medicine particularly as it applies to psychiatry.

Given these developments, the syndromic diagnoses in DSM-5 will in the future likely be replaced by a new diagnostic schema that breaks down existing heterogenous syndromic diagnoses into pathophysiologically and etiologically meaningful entities using insights gained from genetic and biomarker data as well as functional brain imaging. Theoretically, those insights will lead to new modalities of treatment, including somatic treatments that target novel mechanisms of action, coupled to more effective psychosocial therapies—with both therapies guided by diagnostic tests to monitor response to specific treatment interventions.

During this transition from the past to the future, answers to the questions I’ve posed here about laboratory testing in psychiatry will, I hope, help the practitioner understand, evaluate, and incorporate these changes readily into practice.

 


Disclosure
The author serves on the scientific advisory board of Assurex Health, the developer and marketer of the GeneSight test.
References


1. Preskorn SH, Biggs JT. Use of tricyclic antidepressant blood levels. N Engl J Med. 1978;298(3):166.
2. Schildkraut JJ. Biogenic amines and affective disorders. Annu Rev Med. 1974;25(0):333-348.
3. Maas JW. Biogenic amines and depression. Biochemical and pharmacological separation of two types of depression. Arch Gen Psychiatry. 1975;32(11):1357-1361.
4. Carroll BJ, Feinberg M, Greden JF, et al. A specific laboratory test for the diagnosis of melancholia. Standardization, validation, and clinical utility. Arch Gen Psychiatry. 1981;38(1):15-22.
5. Wehler C, Preskorn S. High false-positive rate of a putative biomarker test to aid in the diagnosis of schizophrenia. J Clin Psychiatry. In press.
6. Savitz J, Preskorn S, Teague TK, et al. Minocycline and aspirin in the treatment of bipolar depression: a protocol for a proof-of-concept, randomised, double-blind, placebo-controlled, 2x2 clinical trial. BMJ Open. 2012;2(1):e000643. doi: 10.1136/bmjopen-2011-000643.
7. Rogers HL, Bhattaram A, Zineh I, et al. CYP2D6 genotype information to guide pimozide treatment in adult and pediatric patients: basis for the U.S. Food and Drug Administration’s new dosing recommendations. J Clin Psychiatry. 2012;73(9):1187-1190.
8. Potkin S, Preskorn S, Hochfeld M, et al. A thorough QTc study of 3 doses of iloperidone including metabolic inhibition via CYP2D6 and/or CYP3A4 inhibition and a comparison to quetiapine and ziprasidone. J Clin Psychopharmacol. 2013;33(1):3-10.
9. Howland RH. Pharmacogenetic testing in psychiatry: not (quite) ready for primetime. J Psychosoc Nurs Ment Health Serv. 2014;52(11):13-16.
10. Rosenblat JD, Lee Y, McIntyre RS. Does pharmacogenomics testing improve clinical outcomes for major depressive disorder? A systematic review of clinical trials and cost-effectiveness studies. J Clin Psychiatry. In press.
11. Nassan M, Nicholson WT, Elliott MA, et al. Pharmacokinetic pharmacogenetic prescribing guidelines for antidepressants: a template for psychiatric precision medicine. Mayo Clin Proc. In press.
12. Altar CA, Carhart JM, Allen JD, et al. Clinical validity: combinatorial pharmacogenomics predicts antidepressant responses and healthcare utilizations better than single gene phenotypes. Pharmacogenomics J. 2015;15(5):443-451.
13. Preskorn S, Kane C, Lobello K, et al. Cytochrome P450 2D6 phenoconversion is common in patients being treated for depression: implications for personalized medicine. J Clin Psychiatry. 2013;74(6):614-621.
14. Niculescu AB, Levey DF, Phalen PL, et al. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol Psychiatry. 2015;20(11):1266-1285.

References


1. Preskorn SH, Biggs JT. Use of tricyclic antidepressant blood levels. N Engl J Med. 1978;298(3):166.
2. Schildkraut JJ. Biogenic amines and affective disorders. Annu Rev Med. 1974;25(0):333-348.
3. Maas JW. Biogenic amines and depression. Biochemical and pharmacological separation of two types of depression. Arch Gen Psychiatry. 1975;32(11):1357-1361.
4. Carroll BJ, Feinberg M, Greden JF, et al. A specific laboratory test for the diagnosis of melancholia. Standardization, validation, and clinical utility. Arch Gen Psychiatry. 1981;38(1):15-22.
5. Wehler C, Preskorn S. High false-positive rate of a putative biomarker test to aid in the diagnosis of schizophrenia. J Clin Psychiatry. In press.
6. Savitz J, Preskorn S, Teague TK, et al. Minocycline and aspirin in the treatment of bipolar depression: a protocol for a proof-of-concept, randomised, double-blind, placebo-controlled, 2x2 clinical trial. BMJ Open. 2012;2(1):e000643. doi: 10.1136/bmjopen-2011-000643.
7. Rogers HL, Bhattaram A, Zineh I, et al. CYP2D6 genotype information to guide pimozide treatment in adult and pediatric patients: basis for the U.S. Food and Drug Administration’s new dosing recommendations. J Clin Psychiatry. 2012;73(9):1187-1190.
8. Potkin S, Preskorn S, Hochfeld M, et al. A thorough QTc study of 3 doses of iloperidone including metabolic inhibition via CYP2D6 and/or CYP3A4 inhibition and a comparison to quetiapine and ziprasidone. J Clin Psychopharmacol. 2013;33(1):3-10.
9. Howland RH. Pharmacogenetic testing in psychiatry: not (quite) ready for primetime. J Psychosoc Nurs Ment Health Serv. 2014;52(11):13-16.
10. Rosenblat JD, Lee Y, McIntyre RS. Does pharmacogenomics testing improve clinical outcomes for major depressive disorder? A systematic review of clinical trials and cost-effectiveness studies. J Clin Psychiatry. In press.
11. Nassan M, Nicholson WT, Elliott MA, et al. Pharmacokinetic pharmacogenetic prescribing guidelines for antidepressants: a template for psychiatric precision medicine. Mayo Clin Proc. In press.
12. Altar CA, Carhart JM, Allen JD, et al. Clinical validity: combinatorial pharmacogenomics predicts antidepressant responses and healthcare utilizations better than single gene phenotypes. Pharmacogenomics J. 2015;15(5):443-451.
13. Preskorn S, Kane C, Lobello K, et al. Cytochrome P450 2D6 phenoconversion is common in patients being treated for depression: implications for personalized medicine. J Clin Psychiatry. 2013;74(6):614-621.
14. Niculescu AB, Levey DF, Phalen PL, et al. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol Psychiatry. 2015;20(11):1266-1285.

Issue
Current Psychiatry - 15(4)
Issue
Current Psychiatry - 15(4)
Page Number
19-22,58
Page Number
19-22,58
Publications
Publications
Topics
Article Type
Display Headline
Genetic and related laboratory tests in psychiatry: What mental health practitioners need to know
Display Headline
Genetic and related laboratory tests in psychiatry: What mental health practitioners need to know
Legacy Keywords
genetics, therapeutic drug monitoring, pharmacogenomic, immunoassay
Legacy Keywords
genetics, therapeutic drug monitoring, pharmacogenomic, immunoassay
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Protect against drug-drug interactions with anxiolytics

Article Type
Changed
Tue, 12/11/2018 - 15:09
Display Headline
Protect against drug-drug interactions with anxiolytics

Patients with anxiety disorders are at risk for drug-drug interactions (DDIs) with anxiolytics because they often take medications for comorbid medical or psychiatric illnesses.1-3 Prescribing anxiolytics for them without contemplating both physiology and chemistry leads to what Osler called “popgun pharmacy, hitting now the malady and again the patient,” while “not knowing which.”4

To help you “hit” the anxiety instead of the patient,1 we explain the pharmacokinetics and pharmacodynamics of benzodiazepines, buspirone, and propranolol. Practical tables provide information at a glance about which combinations to avoid and which have potential clinical effects (Box 1) you could use to your patients’ advantage.

Box

3 variables explain a drug’s clinical effect

Clinical effect=Affinity for site of action (pharmacodynamics)×Concentration at site of action (pharmacokinetics)×Patient’s biology (genetics, age, disease, internal environment)

Pharmacodynamics

What a drug does to the body (actions that mediate its efficacy and adverse effects)

Pharmacokinetics

What the body does to a drug (absorption, distribution, metabolism, elimination) that determines its concentration at the site of action

Patient’s biology

Why patients respond differently to the same dose of the same medication (internal environment includes what patients consume, such as foods and co-prescribed drugs)

Benzodiazepines

Benzodiazepines provide an anxiolytic effect by increasing the relative efficiency of the gamma-aminobutyric acid (GABA) receptor when it is stimulated by GABA.5 As a class, benzodiazepines are efficacious for treating panic disorder, social anxiety disorder, generalized anxiety disorder, alcohol withdrawal, and situational anxiety.

Oxidative metabolism. Some benzodiazepines require bio-transformation in the liver by oxidative metabolism; others—such as lorazepam, oxazepam, and emazepam—undergo only glucuronidation reactions and do not have active metabolites (Table 1).6-8

Table 1

Benzodiazepines: How metabolized and half-lives

BenzodiazepineMetabolismHalf-life (includes metabolites)
AlprazolamOxidation 3A3/48 to 12 hrs
ChlordiazepoxideOxidation 3A3/410 to 20 hrs
ClonazepamOxidation 3A3/418 to 50 hrs
ClorazepateOxidation 3A3/440 to 100 hrs
DiazepamOxidation 1A2, 2C8/9, 2C19, 3A3/420 to 70 hrs
LorazepamConjugation10 to 20 hrs
OxazepamConjugation5 to 15 hrs
Source: References 5-7.
Diazepam is a classic example of the first group; its oxidative metabolism is mediated by cytochrome P-450 (CYP) enzymes 1A2, 2C8/9, 2D19, and 3A3/4. Others in this group—alprazolam, clonazepam, midazolam, and triazolam—depend on CYP 3A3/4 for oxidative metabolism.

Benzodiazepines that undergo oxidative metabolism are more likely than those that do not to be influenced by old age, liver disease, or co-administration of other drugs that increase or decrease hepatic CYP enzyme function. Some (midazolam and triazolam) have high first-pass metabolism before reaching systemic circulation.

Pharmacodynamic DDIs. Giving benzodiazepines with other CNS depressants—such as barbiturates, tricyclics and tetracyclics, dopamine receptor antagonists, opioids, or antihistamines, or alcohol—can cause potentially serious oversedation and respiratory depression (Table 2).

Table 2

Clinical effects of drug-drug interactions with benzodiazepines

Pharmacodynamic
Respiratory depression with alcohol, barbiturates, tricyclic and tetracyclic drugs, dopamine receptor antagonists, opioids, antihistamines
With mirtazapine ↑ sedation
With lithium, antipsychotics, and clonazepam → ataxia and dysarthria
With clozapine → delirium
Pharmacokinetic
Cimetidine, disulfiram, isoniazid, estrogen, oral contraceptives ↑ diazepam, chlordiazepoxide plasma concentrations
Nefazodone and fluvoxamine ↑ plasma concentration of triazolam, alprazolam
Carbamazepine ↓ alprazolam plasma concentration
Food, antacids ↓ benzodiazepine plasma concentrations
Cigarette smoking ↑ benzodiazepine metabolism
Benzodiazepines ↑ plasma concentrations of digoxin, phenytoin
Patients with anxiety disorders may use alcohol to self-medicate their anxiety, especially in social situations. Acute and chronic alcohol use with psychotropics may trigger toxic interactions, including fatal poisoning. Combining benzodiazepines with alcohol, opioids, or mirtazapine potentiates sedation through central H-1 antagonism and GABA promotion.2 Acute alcohol ingestion also delays the oxidative metabolism of many drugs.9

Using benzodiazepines with lithium or antipsychotics may cause ataxia and dysarthria, and benzodiazepines with clozapine can cause delirium.

At-risk patients. Benzodiazepine use is a significant predictor of falling, especially in elderly persons taking more than one sedative. In a controlled study of hospitalized older patients, 84 (46%) of 181 who fell were taking one or more benzodiazepine, compared with 48 (27%) of 181 age-matched controls who did not fall.10 The message: seek an alternative to benzodiazepines to sedate older patients, especially those taking another CNS depressant.

Alprazolam and DDIs. Alprazolam is commonly prescribed, despite its high potential for abuse and association with dangerous DDIs:

  • A study of 172 deaths involving oxycodone showed that 117 patients died from combined drug toxicity. Benzodiazepines (detected in 96 cases) were the most common co-intoxicants and were led by alprazolam.11
  • Benzodiazepine abuse is common among clients at methadone maintenance clinics and was reported in 3 fatal drug overdoses caused by co-ingestion of methadone and alprazolam.12
  • Cocaine and methadone were the most common co-intoxicants with alprazolam in a study of 87 deaths attributed to combined drug toxicity.13
  • In a study of patients who overdosed with benzodiazepines, 22% of those who took alprazolam required ICU admission. This was twice the rate of ICU admission after overdose with other benzodiazepines.14
 

 

These studies indicate that alprazolam may be more toxic than other benzodiazepines in overdose and when used with other drugs. We recommend that you exercise great care when prescribing alprazolam, particularly for patients who may be at risk of deliberate self-poisoning and lethal DDIs.

Pharmacokinetic DDIs. Diazepam and chlordiazepoxide plasma concentrations increase in combination with drugs that inhibit CYP enzymes, including cimetidine, disulfiram, isoniazid, estrogen, and oral contraceptives.15

Nefazodone—a CYP 3A3/4 inhibitor—can increase plasma concentrations of triazolam and alprazolam to potentially toxic levels. Nefazodone’s manufacturer recommends lowering triazolam dosages by 75% and alprazolam dosages by 50% when used with nefazodone.3

Carbamazepine—a CYP 3A3/4 inducer—induces both its own and other drugs’ metabolism. It can lower plasma concentrations of alprazolam, clonazepam, midazolam, and triazolam, which are metabolized by 3A3/4. Smoking, food, and antacids also may decrease benzodiazepine plasma concentrations.

As perpetuator drugs, benzodiazepines might increase digoxin plasma concentration, probably because of reduced digoxin renal clearance.16 Diazepam may inhibit CYP 2C9 and/or 2C19 by being an alternate substrate for enzymebinding sites,15,17 increasing the concentration of other drugs such as phenytoin.

Buspirone: Complicated pharmacology

One of buspirone’s major clinical advantages is that it does not pharmacodynamically or pharmacokinetically affect benzodiazepines. Buspirone, the only azaspirodecanedione marketed in the United States, has complex central 5-HT effects.18,19 Because it is a partial 5-HT1A agonist, buspirone’s net effect depends on 5-HT concentration at the receptor:

  • If 5-HT concentration is low, buspirone will act as an agonist.
  • If 5-HT concentration is high, buspirone—being a partial agonist—will antagonize the effect of excessive 5-HT.
Buspirone also acts at postsynaptic and presynaptic 5-HT1A receptors, which mediate different physiologic mechanisms in the brain. Finally, buspirone may act more as a full agonist at postsynaptic than at presynaptic 5-HT1A receptors.20

Buspirone’s pharmacology is further complicated by its conversion via oxidative metabolism into an active metabolite—1-pheyl-piperazine (1-PP). Buspirone is a CYP 3A3/4 enzyme substrate, so it is extensively metabolized as it crosses the duodenum and passes through the liver. As a result, the parent drug has low bioavailability and is principally converted into 1-PP before entering systemic circulation.6

1-PP works differently than the parent drug. As an alpha-2-adrenergic antagonist, 1-PP increases the firing rate of adrenergic neurons in the locus ceruleus by blocking a receptor in presynaptic feedback system.

Which traits of buspirone and its active metabolite produce the drug’s anxiolytic effect? It might be one of these, all of them, or some other unknown trait.

Pharmacodynamic DDIs. Presumably because of its effects on serotonin release at 5-HT1A receptors, buspirone may cause hypertensive episodes when used with monoamine oxidase inhibitors (MAOIs) (Table 3). This is why a 2-week washout is recommended between discontinuing an MAOIs and starting buspirone.21

Table 3

Clinical effects of drug-drug interactions with buspirone

Pharmacodynamic
DO NOT use buspirone with monoamine oxidase inhibitors (MAOIs); allow 2-week washout after stopping an MAOI before starting buspirone
Pharmacokinetic
Food ↑ buspirone Cmax and AUC 2-fold
Renal impairment ↑ buspirone plasma concentration 2-fold
Hepatic impairment ↑ buspirone Cmax and AUC 15-fold and ↑ half-life 2-fold
Verapamil, diltiazem, erythromycin, itraconazole ↑ buspirone plasma concentration
Rifampicin ↓ buspirone plasma concentration 10-fold
Buspirone ↑ haloperidol plasma concentration
Erythromycin, itraconazole, nefazodone, grapefruit juice ↑ buspirone plasma concentration
Cmax: maximum drug concentration
AUC: area under the curve (mathematical calculation of the body’s total exposure to a drug over time)
In theory, buspirone might cause serotonin syndrome when combined with MAOIs. Rare cases of serotonin syndrome have been reported in patients taking buspirone and selective serotonin reuptake inhibitors (SSRIs) and/or trazodone.6 On the other hand, using buspirone to augment SSRIs can cause therapeutic DDIs. Some researchers have added buspirone when patients have not benefited from SSRI monotherapy because:

  • buspirone affects 5-HT mechanisms
  • drugs that affect serotonin reuptake inhibition, 5HT1A receptors, and 5HT2 receptors may have synergy.20
Pharmacokinetic DDIs. Avoid combining buspirone with verapamil, diltiazem, erythromycin, or itraconazole because competitive enzyme inhibition will substantially increase buspirone’s plasma concentration.21

Some SSRIs—such as high-dose fluoxetine and usual doses of fluvoxamine—may increase buspirone serum concentration by inhibiting CYP 3A4.6 Consider this clinical effect before you combine an SSRI with buspirone. Using buspirone with fluoxetine, paroxetine, or bupropion also increases serum 1-PP. This increase, which occurs when CYP 2D6 slows 1-PP clearance, could cause euphoria, mania, or seizures.20

Coadministering rifampin can lower buspirone plasma concentrations almost 10-fold because rifampin induces CYP 3A3/4.22

As a perpetuator, buspirone can increase haloperidol plasma concentrations, but probably not to a clinically important extent. In an open trial, Goff23 added buspirone, mean dosage 23.8 mg/d, to a stable regimen of haloperidol in 7 patients with schizophrenia. Although haloperidol’s mean plasma concentration increased by 26% after 6 weeks, this modest change would be difficult to detect in clinical practice.

 

 

Huang et al24 found no clinically significant pharmacokinetic interaction between buspirone, 10 mg tid, and haloperidol, 10 to 40 mg/d, during 6 weeks of coadministration in 27 patients with schizophrenia.

Propranolol: Beta-blocking anxiolytic

Propranolol is prescribed off-label for anxiety disorders more often than other beta blockers. It may help patients with situational or performance anxiety.

Beta-adrenergic blockers competitively antagonize norepinephrine and epinephrine at the beta-adrenergic receptor. These cardiovascular agents can reduce many of anxiety’s peripheral manifestations, such as tachycardia, diaphoresis, trembling, and blushing. All beta blockers share this pharmacologic effect, but their pharmacokinetics differ greatly. Some depend on a single CYP enzyme for clearance (metoprolol, by CYP 2D6), whereas others, such as propranolol, are metabolized by multiple CYP enzymes.

Pharmacodynamic DDIs. Drugs that block alpha-1 adrenergic receptors potentiate beta blockers’ blood pressure-lowering effects and increase the risk of orthostatic hypotension. This is probably why haloperidol can potentiate propranolol’s hypotensive effects.6 Other alpha-1 adrenergic antagonists—though not normally classified as such—include some tertiary amine tricyclic antidepressants (amitriptyline and imipramine) and some antipsychotics (quetiapine).

Reports have associated hypertensive crises and bradycardia with coadministration of beta blockers and MAOIs.21 Depressed myocardial contractility and A-V nodal conduction may occur when beta blockers are combined with calcium channel inhibitors.21 Beta blockers also can decrease IV anesthetic dose requirements because they decrease cardiac output.25

In patients using insulin for diabetes mellitus, propranolol inhibits recovery from insulin-induced hypoglycemia and may cause hypertension and bradycardia. Beta blockers also can mask the tachycardia that usually accompanies insulin-induced hypoglycemia.

Pharmacokinetic DDIs. Propranolol has an extensive first-pass effect, being etabolized in the liver to active and inactive compounds that interact with CYP enzymes 1A2, 2C18, 2C19 and 2D6.6

Coadministering strong CYP 2D6 inhibitors such as bupropion, fluoxetine, or paroxetine can reduce propanolol clearance, increasing its effect and risking cardiac toxicity6 (Table 4). CYP 1A2 inhibitors such as amiodarone and fluoroquinolones or CYP 2C19 inhibitors such as fluvoxamine also increase serum concentrations of propranolol.

Table 4

How to avoid drug interactions with three common anxiolytics*

When prescribing benzodiazepines…
DODO NOT
Advise patients not to combine benzodiazepines with alcoholUse with other CNS depressants or nefazodone
Talk to patients about potential for abuse/dependency, and monitor benzodiazepine useUse in elderly patients or in patients with high potential for substance abuse
When prescribing buspirone…
DODO NOT
Allow a 2-week washout between discontinuing an MAOI and starting buspironeUse with MAOIs, verapamil, diltiazem, erythromycin, or itraconazole
Consider adding buspirone when SSRI monotherapy has not adequately helped patients with anxietyCo-administer with rifampin
Combine with benzodiazepines, if needed 
When prescribing propranolol…
DODO NOT
Educate patients using insulin for diabetes mellitus that propranolol may inhibit recovery from insulin-induced hypoglycemia, cause bradycardia, or mask tachycardiaCombine with medications with strong hypotensive effects
 Coadminister with strong CYP 2D6 or 1A2 inhibitors
Recheck anticonvulsant plasma concentrations after starting propranololAdd to calcium inhibitors for patients with ↓ myocardial contractility and A-V nodal conduction
* Before prescribing any anxiolytic, review all co-prescribed medications for potential DDIs
DDI: drug-drug interaction
MAOI: monoamine oxidase inhibitor
SSRI: selective serotonin reuptake inhibitor
On the other hand, CYP inducers such as barbiturates, phenytoin, and cigarette smoking can increase propranolol elimination and decrease its serum levels.26 Hyperthyroidism may enhance propranolol’s presystemic clearance but has little effect on its half life.27

As a perpetuator, propranolol produces small increases in diazepam concentration, suggesting that the beta-blocker inhibits diazepam metabolism. This interaction can impair kinetic visual acuity, which is correlated with the ability to discriminate moving objects in space.26

Propranolol increases plasma concentrations of antipsychotics, anticonvulsants, theophylline, and levothyroxine (Table 5)—possibly because of the beta blocker’s negative inotropic effects (decreased cardiac output reduces hepatic and renal blood flow).

Table 5

Clinical effects of drug-drug interactions with propranolol

Pharmacodynamic
With MAO inhibitors → hypertensive crisis and bradycardia
With calcium channel inhibitors → ↓ myocardial contractility and A-V nodal conduction
↓ intravenous anesthetic dose requirements
↓ diazepam metabolism
↓ median effective dosage of valproate and diazepam; might improve antiepileptic potential of valproate
Pharmacokinetic
↑ plasma concentration of antipsychotics, anticonvulsants, theophylline, levothyroxine
Barbiturates, phenytoin, and cigarette smoking ↑ propranolol elimination
Related resources

Drug brand names

  • Alprazolam • Xanax
  • Bupropion • Wellbutrin
  • Buspirone • BuSpar
  • Carbamazepine • Carbatrol, others
  • Chlordiazepoxide • Librium
  • Cimetidine • Tagamet
  • Clonazepam • Klonopin
  • Clorazepate • Tranxene
  • Clozapine • Clozaril
  • Diazepam • Valium
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Itraconazole • Sporanox
  • Lorazepam • Ativan
  • Midazolam • Versed
  • Mirtazapine • Remeron
  • Oxazepam • Serax
  • Paroxetine • Paxil
  • Phenytoin • Dilantin
  • Propranolol • Inderal
  • Quetiapine • Seroquel
  • Rifampin • Rifadin, Rimactane
  • Triazolam • Halcion
  • Valproate • various
  • Verapamil • Calan, Isoptin
Disclosures

Drs. Ramadan and Werder report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

 

 

Dr. Preskorn has received grants or has been a consultant or speaker for Abbott Laboratories, AstraZeneca Pharmaceuticals, Boehringer-Ingelheim, Bristol-Myers Squibb Co., Merck & Co., Eisai, Eli Lilly and Co., GlaxoSmithKline, Janssen Pharmaceutica, Johnson & Johnson, Novartis Pharmaceuticals Corp., Organon, Otsuka America Pharmaceutical, Pfizer, Solvay Pharmaceuticals, Sanofi-Aventis, and Wyeth.

References

1. Preskorn S, Flockhart D. Psychiatric drug interactions guide. New York: MBL Communications.; 2004.

2. Bruce SE, Yonkers KA, Otto MW, et al. Influence of psychiatric comorbidity on recovery and recurrence in generalized anxiety disorder, social phobia, and panic disorder 12-year prospective study. Am J Psychiatry 2005;162:1179-87.

3. Nemeroff CB. Use of atypical antipsychotics in refractory depression and anxiety. J Clin Psychiatry 2005;66(Suppl 8):13-21.

4. Bean RB, Bean WB. Sir William Osler: Aphorisms from his bedside teaching and writing. Springfield, IL: Charles C. Thomas; 1961:53.

5. Tasman A, Kay J, Lieberman JA. Psychiatry Therapeutics. 2nd ed. West Sussex, UK: John Wiley & Sons; 2003:347.

6. Fuller M, Sajatovic M. Drug information for mental health. 3rd ed. Hudson, OH: Lexi-Comp; 2001.

7. Stahl SM. Essential psychopharmacology: Neuroscientific basis and practical applications. 2nd ed. New York: Cambridge University Press; 2000.

8. Janicak PG, Davis JM, Preskorn SH, Ayad FJ. Principles and practice of psychopharmacology. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2001.

9. Tanaka E. Toxicological interactions involving psychiatric drugs and alcohol: an update. J Clin Pharm Ther 2003;28(2):81-95.

10. Frels C, Williams P, Narayanan S, Gariballa SE. Iatrogenic causes of falls in hospitalised elderly patients: a case-control study. Postgrad Med 2002;78(922):487-9.

11. Wolf BC, Lavezzi WA, Sullivan LM, Flannagan LM. One hundred seventy two deaths involving the use of oxycodone in Palm Beach County. J Forensic Sci 2005;50(1):192-5.

12. Rogers WO, Hall MA, Brissie RM, Robinson CA. Detection of alprazolam in three cases of methadone/benzodiazepine overdose. J Forensic Sci 1997;42(1):155-6.

13. Wolf BC, Lavezzi WA, Sullivan LM, et al. Alprazolam-related deaths in Palm Beach County. Am J Forensic Med Pathol 2005;26(1):24-7.

14. Isbister GK, O’Regan L, Sibbritt D, et al. Alprazolam is relatively more toxic than other benzodiazepines in overdose. Br J Clin Pharmacol 2004;58(1):88-95.

15. Sadock BJ, Sadock VA. Kaplan and Sadock’s pocket handbook of psychiatric drug treatment. 3rd ed. Philadelphia: Lippincott Williams & Wilkins; 2001.

16. Tollefson G, Lesar T, Grothe D, et al. Alprazolam-related digoxin toxicity. Am J Psychiatry 1984;141(12):1612-3.

17. Murphy A, Wilbur K. Phenytoin-diazepam interaction. Ann Pharmacother 2003;37(5):659-3.

18. Sharp T, McQuade R, Bramwell S, et al. Effect of acute and repeated administration of 5-HT1A receptor agonists on 5-HT release in rat brain in vivo. Naunhyn Schmiedebergs Arch Pharmacol 1993;348(4):339-46.

19. Van den Hooff P, Galvan M. Actions of 5-hydroxytryptamine and 5-HT1A receptor ligands on rat dorso-lateral septal neurons in vitro. Br J Pharmacol 1992;106(4):893-9.

20. Preskorn SH Do you believe in magic? Journal of Practical Psychiatry and Behavioral Health March 1997;99-103

21. Physicians’ Desk Reference. 59th ed. Montvale, NJ: Thomson PDR; 2005.

22. Mahmood I, Sahajwalla C. Clinical pharmacokinetics and pharmacodynamics of buspirone, an anxiolytic drug. Clin Pharmacokinet 1999;36(4):277-87.

23. Goff DC, Midha KK, Brotman AW, et al. An open trial of buspirone added to neuroleptics in schizophrenic patients. J Clin Psychopharmacol 1991;11(3):193-7.

24. Huang HF, Jann MW, Wei FC, et al. Lack of pharmacokinetic interaction between buspirone and haloperidol in patients with schizophrenia. J Clin Pharmacol 1996;36(10):963-9.

25. Avram MJ, Krejcie TC, Henthorn TK, et al. Beta-adrenergic blockade affects initial drug distribution due to decreased cardiac output and altered blood flow distribution. J Pharmacol Exp Ther 2004;311(2):617-24.

26. Hawksworth G, Betts T, Crowe A, et al. Diazepam/beta-adrenoceptor antagonist interactions. Br J Clin Pharmacol 1984;17(Suppl 1):69S-76S.

27. Hallengren B, Nilsson OR, Karlberg BE, et al. Influence of hyperthyroidism on the kinetics of methimazole, propranolol, metoprolol and atenolol. Eur J Clin Pharmacol 1982;21(5):379-84.

Article PDF
Author and Disclosure Information

Mohamed I. Ramadan, MD
Staff psychiatrist, Mohave Mental Health Clinic, Kingman, AZ
Steve F. Werder, DO
Assistant professor, Department of psychiatry and behavioral sciences, University of Kansas, School of Medicine, Wichita

Sheldon H. Preskorn, MD
Chairman, Department of psychiatry and behavioral sciences, University of Kansas, School of Medicine, Wichita

Issue
Current Psychiatry - 05(05)
Publications
Page Number
16-28
Sections
Author and Disclosure Information

Mohamed I. Ramadan, MD
Staff psychiatrist, Mohave Mental Health Clinic, Kingman, AZ
Steve F. Werder, DO
Assistant professor, Department of psychiatry and behavioral sciences, University of Kansas, School of Medicine, Wichita

Sheldon H. Preskorn, MD
Chairman, Department of psychiatry and behavioral sciences, University of Kansas, School of Medicine, Wichita

Author and Disclosure Information

Mohamed I. Ramadan, MD
Staff psychiatrist, Mohave Mental Health Clinic, Kingman, AZ
Steve F. Werder, DO
Assistant professor, Department of psychiatry and behavioral sciences, University of Kansas, School of Medicine, Wichita

Sheldon H. Preskorn, MD
Chairman, Department of psychiatry and behavioral sciences, University of Kansas, School of Medicine, Wichita

Article PDF
Article PDF

Patients with anxiety disorders are at risk for drug-drug interactions (DDIs) with anxiolytics because they often take medications for comorbid medical or psychiatric illnesses.1-3 Prescribing anxiolytics for them without contemplating both physiology and chemistry leads to what Osler called “popgun pharmacy, hitting now the malady and again the patient,” while “not knowing which.”4

To help you “hit” the anxiety instead of the patient,1 we explain the pharmacokinetics and pharmacodynamics of benzodiazepines, buspirone, and propranolol. Practical tables provide information at a glance about which combinations to avoid and which have potential clinical effects (Box 1) you could use to your patients’ advantage.

Box

3 variables explain a drug’s clinical effect

Clinical effect=Affinity for site of action (pharmacodynamics)×Concentration at site of action (pharmacokinetics)×Patient’s biology (genetics, age, disease, internal environment)

Pharmacodynamics

What a drug does to the body (actions that mediate its efficacy and adverse effects)

Pharmacokinetics

What the body does to a drug (absorption, distribution, metabolism, elimination) that determines its concentration at the site of action

Patient’s biology

Why patients respond differently to the same dose of the same medication (internal environment includes what patients consume, such as foods and co-prescribed drugs)

Benzodiazepines

Benzodiazepines provide an anxiolytic effect by increasing the relative efficiency of the gamma-aminobutyric acid (GABA) receptor when it is stimulated by GABA.5 As a class, benzodiazepines are efficacious for treating panic disorder, social anxiety disorder, generalized anxiety disorder, alcohol withdrawal, and situational anxiety.

Oxidative metabolism. Some benzodiazepines require bio-transformation in the liver by oxidative metabolism; others—such as lorazepam, oxazepam, and emazepam—undergo only glucuronidation reactions and do not have active metabolites (Table 1).6-8

Table 1

Benzodiazepines: How metabolized and half-lives

BenzodiazepineMetabolismHalf-life (includes metabolites)
AlprazolamOxidation 3A3/48 to 12 hrs
ChlordiazepoxideOxidation 3A3/410 to 20 hrs
ClonazepamOxidation 3A3/418 to 50 hrs
ClorazepateOxidation 3A3/440 to 100 hrs
DiazepamOxidation 1A2, 2C8/9, 2C19, 3A3/420 to 70 hrs
LorazepamConjugation10 to 20 hrs
OxazepamConjugation5 to 15 hrs
Source: References 5-7.
Diazepam is a classic example of the first group; its oxidative metabolism is mediated by cytochrome P-450 (CYP) enzymes 1A2, 2C8/9, 2D19, and 3A3/4. Others in this group—alprazolam, clonazepam, midazolam, and triazolam—depend on CYP 3A3/4 for oxidative metabolism.

Benzodiazepines that undergo oxidative metabolism are more likely than those that do not to be influenced by old age, liver disease, or co-administration of other drugs that increase or decrease hepatic CYP enzyme function. Some (midazolam and triazolam) have high first-pass metabolism before reaching systemic circulation.

Pharmacodynamic DDIs. Giving benzodiazepines with other CNS depressants—such as barbiturates, tricyclics and tetracyclics, dopamine receptor antagonists, opioids, or antihistamines, or alcohol—can cause potentially serious oversedation and respiratory depression (Table 2).

Table 2

Clinical effects of drug-drug interactions with benzodiazepines

Pharmacodynamic
Respiratory depression with alcohol, barbiturates, tricyclic and tetracyclic drugs, dopamine receptor antagonists, opioids, antihistamines
With mirtazapine ↑ sedation
With lithium, antipsychotics, and clonazepam → ataxia and dysarthria
With clozapine → delirium
Pharmacokinetic
Cimetidine, disulfiram, isoniazid, estrogen, oral contraceptives ↑ diazepam, chlordiazepoxide plasma concentrations
Nefazodone and fluvoxamine ↑ plasma concentration of triazolam, alprazolam
Carbamazepine ↓ alprazolam plasma concentration
Food, antacids ↓ benzodiazepine plasma concentrations
Cigarette smoking ↑ benzodiazepine metabolism
Benzodiazepines ↑ plasma concentrations of digoxin, phenytoin
Patients with anxiety disorders may use alcohol to self-medicate their anxiety, especially in social situations. Acute and chronic alcohol use with psychotropics may trigger toxic interactions, including fatal poisoning. Combining benzodiazepines with alcohol, opioids, or mirtazapine potentiates sedation through central H-1 antagonism and GABA promotion.2 Acute alcohol ingestion also delays the oxidative metabolism of many drugs.9

Using benzodiazepines with lithium or antipsychotics may cause ataxia and dysarthria, and benzodiazepines with clozapine can cause delirium.

At-risk patients. Benzodiazepine use is a significant predictor of falling, especially in elderly persons taking more than one sedative. In a controlled study of hospitalized older patients, 84 (46%) of 181 who fell were taking one or more benzodiazepine, compared with 48 (27%) of 181 age-matched controls who did not fall.10 The message: seek an alternative to benzodiazepines to sedate older patients, especially those taking another CNS depressant.

Alprazolam and DDIs. Alprazolam is commonly prescribed, despite its high potential for abuse and association with dangerous DDIs:

  • A study of 172 deaths involving oxycodone showed that 117 patients died from combined drug toxicity. Benzodiazepines (detected in 96 cases) were the most common co-intoxicants and were led by alprazolam.11
  • Benzodiazepine abuse is common among clients at methadone maintenance clinics and was reported in 3 fatal drug overdoses caused by co-ingestion of methadone and alprazolam.12
  • Cocaine and methadone were the most common co-intoxicants with alprazolam in a study of 87 deaths attributed to combined drug toxicity.13
  • In a study of patients who overdosed with benzodiazepines, 22% of those who took alprazolam required ICU admission. This was twice the rate of ICU admission after overdose with other benzodiazepines.14
 

 

These studies indicate that alprazolam may be more toxic than other benzodiazepines in overdose and when used with other drugs. We recommend that you exercise great care when prescribing alprazolam, particularly for patients who may be at risk of deliberate self-poisoning and lethal DDIs.

Pharmacokinetic DDIs. Diazepam and chlordiazepoxide plasma concentrations increase in combination with drugs that inhibit CYP enzymes, including cimetidine, disulfiram, isoniazid, estrogen, and oral contraceptives.15

Nefazodone—a CYP 3A3/4 inhibitor—can increase plasma concentrations of triazolam and alprazolam to potentially toxic levels. Nefazodone’s manufacturer recommends lowering triazolam dosages by 75% and alprazolam dosages by 50% when used with nefazodone.3

Carbamazepine—a CYP 3A3/4 inducer—induces both its own and other drugs’ metabolism. It can lower plasma concentrations of alprazolam, clonazepam, midazolam, and triazolam, which are metabolized by 3A3/4. Smoking, food, and antacids also may decrease benzodiazepine plasma concentrations.

As perpetuator drugs, benzodiazepines might increase digoxin plasma concentration, probably because of reduced digoxin renal clearance.16 Diazepam may inhibit CYP 2C9 and/or 2C19 by being an alternate substrate for enzymebinding sites,15,17 increasing the concentration of other drugs such as phenytoin.

Buspirone: Complicated pharmacology

One of buspirone’s major clinical advantages is that it does not pharmacodynamically or pharmacokinetically affect benzodiazepines. Buspirone, the only azaspirodecanedione marketed in the United States, has complex central 5-HT effects.18,19 Because it is a partial 5-HT1A agonist, buspirone’s net effect depends on 5-HT concentration at the receptor:

  • If 5-HT concentration is low, buspirone will act as an agonist.
  • If 5-HT concentration is high, buspirone—being a partial agonist—will antagonize the effect of excessive 5-HT.
Buspirone also acts at postsynaptic and presynaptic 5-HT1A receptors, which mediate different physiologic mechanisms in the brain. Finally, buspirone may act more as a full agonist at postsynaptic than at presynaptic 5-HT1A receptors.20

Buspirone’s pharmacology is further complicated by its conversion via oxidative metabolism into an active metabolite—1-pheyl-piperazine (1-PP). Buspirone is a CYP 3A3/4 enzyme substrate, so it is extensively metabolized as it crosses the duodenum and passes through the liver. As a result, the parent drug has low bioavailability and is principally converted into 1-PP before entering systemic circulation.6

1-PP works differently than the parent drug. As an alpha-2-adrenergic antagonist, 1-PP increases the firing rate of adrenergic neurons in the locus ceruleus by blocking a receptor in presynaptic feedback system.

Which traits of buspirone and its active metabolite produce the drug’s anxiolytic effect? It might be one of these, all of them, or some other unknown trait.

Pharmacodynamic DDIs. Presumably because of its effects on serotonin release at 5-HT1A receptors, buspirone may cause hypertensive episodes when used with monoamine oxidase inhibitors (MAOIs) (Table 3). This is why a 2-week washout is recommended between discontinuing an MAOIs and starting buspirone.21

Table 3

Clinical effects of drug-drug interactions with buspirone

Pharmacodynamic
DO NOT use buspirone with monoamine oxidase inhibitors (MAOIs); allow 2-week washout after stopping an MAOI before starting buspirone
Pharmacokinetic
Food ↑ buspirone Cmax and AUC 2-fold
Renal impairment ↑ buspirone plasma concentration 2-fold
Hepatic impairment ↑ buspirone Cmax and AUC 15-fold and ↑ half-life 2-fold
Verapamil, diltiazem, erythromycin, itraconazole ↑ buspirone plasma concentration
Rifampicin ↓ buspirone plasma concentration 10-fold
Buspirone ↑ haloperidol plasma concentration
Erythromycin, itraconazole, nefazodone, grapefruit juice ↑ buspirone plasma concentration
Cmax: maximum drug concentration
AUC: area under the curve (mathematical calculation of the body’s total exposure to a drug over time)
In theory, buspirone might cause serotonin syndrome when combined with MAOIs. Rare cases of serotonin syndrome have been reported in patients taking buspirone and selective serotonin reuptake inhibitors (SSRIs) and/or trazodone.6 On the other hand, using buspirone to augment SSRIs can cause therapeutic DDIs. Some researchers have added buspirone when patients have not benefited from SSRI monotherapy because:

  • buspirone affects 5-HT mechanisms
  • drugs that affect serotonin reuptake inhibition, 5HT1A receptors, and 5HT2 receptors may have synergy.20
Pharmacokinetic DDIs. Avoid combining buspirone with verapamil, diltiazem, erythromycin, or itraconazole because competitive enzyme inhibition will substantially increase buspirone’s plasma concentration.21

Some SSRIs—such as high-dose fluoxetine and usual doses of fluvoxamine—may increase buspirone serum concentration by inhibiting CYP 3A4.6 Consider this clinical effect before you combine an SSRI with buspirone. Using buspirone with fluoxetine, paroxetine, or bupropion also increases serum 1-PP. This increase, which occurs when CYP 2D6 slows 1-PP clearance, could cause euphoria, mania, or seizures.20

Coadministering rifampin can lower buspirone plasma concentrations almost 10-fold because rifampin induces CYP 3A3/4.22

As a perpetuator, buspirone can increase haloperidol plasma concentrations, but probably not to a clinically important extent. In an open trial, Goff23 added buspirone, mean dosage 23.8 mg/d, to a stable regimen of haloperidol in 7 patients with schizophrenia. Although haloperidol’s mean plasma concentration increased by 26% after 6 weeks, this modest change would be difficult to detect in clinical practice.

 

 

Huang et al24 found no clinically significant pharmacokinetic interaction between buspirone, 10 mg tid, and haloperidol, 10 to 40 mg/d, during 6 weeks of coadministration in 27 patients with schizophrenia.

Propranolol: Beta-blocking anxiolytic

Propranolol is prescribed off-label for anxiety disorders more often than other beta blockers. It may help patients with situational or performance anxiety.

Beta-adrenergic blockers competitively antagonize norepinephrine and epinephrine at the beta-adrenergic receptor. These cardiovascular agents can reduce many of anxiety’s peripheral manifestations, such as tachycardia, diaphoresis, trembling, and blushing. All beta blockers share this pharmacologic effect, but their pharmacokinetics differ greatly. Some depend on a single CYP enzyme for clearance (metoprolol, by CYP 2D6), whereas others, such as propranolol, are metabolized by multiple CYP enzymes.

Pharmacodynamic DDIs. Drugs that block alpha-1 adrenergic receptors potentiate beta blockers’ blood pressure-lowering effects and increase the risk of orthostatic hypotension. This is probably why haloperidol can potentiate propranolol’s hypotensive effects.6 Other alpha-1 adrenergic antagonists—though not normally classified as such—include some tertiary amine tricyclic antidepressants (amitriptyline and imipramine) and some antipsychotics (quetiapine).

Reports have associated hypertensive crises and bradycardia with coadministration of beta blockers and MAOIs.21 Depressed myocardial contractility and A-V nodal conduction may occur when beta blockers are combined with calcium channel inhibitors.21 Beta blockers also can decrease IV anesthetic dose requirements because they decrease cardiac output.25

In patients using insulin for diabetes mellitus, propranolol inhibits recovery from insulin-induced hypoglycemia and may cause hypertension and bradycardia. Beta blockers also can mask the tachycardia that usually accompanies insulin-induced hypoglycemia.

Pharmacokinetic DDIs. Propranolol has an extensive first-pass effect, being etabolized in the liver to active and inactive compounds that interact with CYP enzymes 1A2, 2C18, 2C19 and 2D6.6

Coadministering strong CYP 2D6 inhibitors such as bupropion, fluoxetine, or paroxetine can reduce propanolol clearance, increasing its effect and risking cardiac toxicity6 (Table 4). CYP 1A2 inhibitors such as amiodarone and fluoroquinolones or CYP 2C19 inhibitors such as fluvoxamine also increase serum concentrations of propranolol.

Table 4

How to avoid drug interactions with three common anxiolytics*

When prescribing benzodiazepines…
DODO NOT
Advise patients not to combine benzodiazepines with alcoholUse with other CNS depressants or nefazodone
Talk to patients about potential for abuse/dependency, and monitor benzodiazepine useUse in elderly patients or in patients with high potential for substance abuse
When prescribing buspirone…
DODO NOT
Allow a 2-week washout between discontinuing an MAOI and starting buspironeUse with MAOIs, verapamil, diltiazem, erythromycin, or itraconazole
Consider adding buspirone when SSRI monotherapy has not adequately helped patients with anxietyCo-administer with rifampin
Combine with benzodiazepines, if needed 
When prescribing propranolol…
DODO NOT
Educate patients using insulin for diabetes mellitus that propranolol may inhibit recovery from insulin-induced hypoglycemia, cause bradycardia, or mask tachycardiaCombine with medications with strong hypotensive effects
 Coadminister with strong CYP 2D6 or 1A2 inhibitors
Recheck anticonvulsant plasma concentrations after starting propranololAdd to calcium inhibitors for patients with ↓ myocardial contractility and A-V nodal conduction
* Before prescribing any anxiolytic, review all co-prescribed medications for potential DDIs
DDI: drug-drug interaction
MAOI: monoamine oxidase inhibitor
SSRI: selective serotonin reuptake inhibitor
On the other hand, CYP inducers such as barbiturates, phenytoin, and cigarette smoking can increase propranolol elimination and decrease its serum levels.26 Hyperthyroidism may enhance propranolol’s presystemic clearance but has little effect on its half life.27

As a perpetuator, propranolol produces small increases in diazepam concentration, suggesting that the beta-blocker inhibits diazepam metabolism. This interaction can impair kinetic visual acuity, which is correlated with the ability to discriminate moving objects in space.26

Propranolol increases plasma concentrations of antipsychotics, anticonvulsants, theophylline, and levothyroxine (Table 5)—possibly because of the beta blocker’s negative inotropic effects (decreased cardiac output reduces hepatic and renal blood flow).

Table 5

Clinical effects of drug-drug interactions with propranolol

Pharmacodynamic
With MAO inhibitors → hypertensive crisis and bradycardia
With calcium channel inhibitors → ↓ myocardial contractility and A-V nodal conduction
↓ intravenous anesthetic dose requirements
↓ diazepam metabolism
↓ median effective dosage of valproate and diazepam; might improve antiepileptic potential of valproate
Pharmacokinetic
↑ plasma concentration of antipsychotics, anticonvulsants, theophylline, levothyroxine
Barbiturates, phenytoin, and cigarette smoking ↑ propranolol elimination
Related resources

Drug brand names

  • Alprazolam • Xanax
  • Bupropion • Wellbutrin
  • Buspirone • BuSpar
  • Carbamazepine • Carbatrol, others
  • Chlordiazepoxide • Librium
  • Cimetidine • Tagamet
  • Clonazepam • Klonopin
  • Clorazepate • Tranxene
  • Clozapine • Clozaril
  • Diazepam • Valium
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Itraconazole • Sporanox
  • Lorazepam • Ativan
  • Midazolam • Versed
  • Mirtazapine • Remeron
  • Oxazepam • Serax
  • Paroxetine • Paxil
  • Phenytoin • Dilantin
  • Propranolol • Inderal
  • Quetiapine • Seroquel
  • Rifampin • Rifadin, Rimactane
  • Triazolam • Halcion
  • Valproate • various
  • Verapamil • Calan, Isoptin
Disclosures

Drs. Ramadan and Werder report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

 

 

Dr. Preskorn has received grants or has been a consultant or speaker for Abbott Laboratories, AstraZeneca Pharmaceuticals, Boehringer-Ingelheim, Bristol-Myers Squibb Co., Merck & Co., Eisai, Eli Lilly and Co., GlaxoSmithKline, Janssen Pharmaceutica, Johnson & Johnson, Novartis Pharmaceuticals Corp., Organon, Otsuka America Pharmaceutical, Pfizer, Solvay Pharmaceuticals, Sanofi-Aventis, and Wyeth.

Patients with anxiety disorders are at risk for drug-drug interactions (DDIs) with anxiolytics because they often take medications for comorbid medical or psychiatric illnesses.1-3 Prescribing anxiolytics for them without contemplating both physiology and chemistry leads to what Osler called “popgun pharmacy, hitting now the malady and again the patient,” while “not knowing which.”4

To help you “hit” the anxiety instead of the patient,1 we explain the pharmacokinetics and pharmacodynamics of benzodiazepines, buspirone, and propranolol. Practical tables provide information at a glance about which combinations to avoid and which have potential clinical effects (Box 1) you could use to your patients’ advantage.

Box

3 variables explain a drug’s clinical effect

Clinical effect=Affinity for site of action (pharmacodynamics)×Concentration at site of action (pharmacokinetics)×Patient’s biology (genetics, age, disease, internal environment)

Pharmacodynamics

What a drug does to the body (actions that mediate its efficacy and adverse effects)

Pharmacokinetics

What the body does to a drug (absorption, distribution, metabolism, elimination) that determines its concentration at the site of action

Patient’s biology

Why patients respond differently to the same dose of the same medication (internal environment includes what patients consume, such as foods and co-prescribed drugs)

Benzodiazepines

Benzodiazepines provide an anxiolytic effect by increasing the relative efficiency of the gamma-aminobutyric acid (GABA) receptor when it is stimulated by GABA.5 As a class, benzodiazepines are efficacious for treating panic disorder, social anxiety disorder, generalized anxiety disorder, alcohol withdrawal, and situational anxiety.

Oxidative metabolism. Some benzodiazepines require bio-transformation in the liver by oxidative metabolism; others—such as lorazepam, oxazepam, and emazepam—undergo only glucuronidation reactions and do not have active metabolites (Table 1).6-8

Table 1

Benzodiazepines: How metabolized and half-lives

BenzodiazepineMetabolismHalf-life (includes metabolites)
AlprazolamOxidation 3A3/48 to 12 hrs
ChlordiazepoxideOxidation 3A3/410 to 20 hrs
ClonazepamOxidation 3A3/418 to 50 hrs
ClorazepateOxidation 3A3/440 to 100 hrs
DiazepamOxidation 1A2, 2C8/9, 2C19, 3A3/420 to 70 hrs
LorazepamConjugation10 to 20 hrs
OxazepamConjugation5 to 15 hrs
Source: References 5-7.
Diazepam is a classic example of the first group; its oxidative metabolism is mediated by cytochrome P-450 (CYP) enzymes 1A2, 2C8/9, 2D19, and 3A3/4. Others in this group—alprazolam, clonazepam, midazolam, and triazolam—depend on CYP 3A3/4 for oxidative metabolism.

Benzodiazepines that undergo oxidative metabolism are more likely than those that do not to be influenced by old age, liver disease, or co-administration of other drugs that increase or decrease hepatic CYP enzyme function. Some (midazolam and triazolam) have high first-pass metabolism before reaching systemic circulation.

Pharmacodynamic DDIs. Giving benzodiazepines with other CNS depressants—such as barbiturates, tricyclics and tetracyclics, dopamine receptor antagonists, opioids, or antihistamines, or alcohol—can cause potentially serious oversedation and respiratory depression (Table 2).

Table 2

Clinical effects of drug-drug interactions with benzodiazepines

Pharmacodynamic
Respiratory depression with alcohol, barbiturates, tricyclic and tetracyclic drugs, dopamine receptor antagonists, opioids, antihistamines
With mirtazapine ↑ sedation
With lithium, antipsychotics, and clonazepam → ataxia and dysarthria
With clozapine → delirium
Pharmacokinetic
Cimetidine, disulfiram, isoniazid, estrogen, oral contraceptives ↑ diazepam, chlordiazepoxide plasma concentrations
Nefazodone and fluvoxamine ↑ plasma concentration of triazolam, alprazolam
Carbamazepine ↓ alprazolam plasma concentration
Food, antacids ↓ benzodiazepine plasma concentrations
Cigarette smoking ↑ benzodiazepine metabolism
Benzodiazepines ↑ plasma concentrations of digoxin, phenytoin
Patients with anxiety disorders may use alcohol to self-medicate their anxiety, especially in social situations. Acute and chronic alcohol use with psychotropics may trigger toxic interactions, including fatal poisoning. Combining benzodiazepines with alcohol, opioids, or mirtazapine potentiates sedation through central H-1 antagonism and GABA promotion.2 Acute alcohol ingestion also delays the oxidative metabolism of many drugs.9

Using benzodiazepines with lithium or antipsychotics may cause ataxia and dysarthria, and benzodiazepines with clozapine can cause delirium.

At-risk patients. Benzodiazepine use is a significant predictor of falling, especially in elderly persons taking more than one sedative. In a controlled study of hospitalized older patients, 84 (46%) of 181 who fell were taking one or more benzodiazepine, compared with 48 (27%) of 181 age-matched controls who did not fall.10 The message: seek an alternative to benzodiazepines to sedate older patients, especially those taking another CNS depressant.

Alprazolam and DDIs. Alprazolam is commonly prescribed, despite its high potential for abuse and association with dangerous DDIs:

  • A study of 172 deaths involving oxycodone showed that 117 patients died from combined drug toxicity. Benzodiazepines (detected in 96 cases) were the most common co-intoxicants and were led by alprazolam.11
  • Benzodiazepine abuse is common among clients at methadone maintenance clinics and was reported in 3 fatal drug overdoses caused by co-ingestion of methadone and alprazolam.12
  • Cocaine and methadone were the most common co-intoxicants with alprazolam in a study of 87 deaths attributed to combined drug toxicity.13
  • In a study of patients who overdosed with benzodiazepines, 22% of those who took alprazolam required ICU admission. This was twice the rate of ICU admission after overdose with other benzodiazepines.14
 

 

These studies indicate that alprazolam may be more toxic than other benzodiazepines in overdose and when used with other drugs. We recommend that you exercise great care when prescribing alprazolam, particularly for patients who may be at risk of deliberate self-poisoning and lethal DDIs.

Pharmacokinetic DDIs. Diazepam and chlordiazepoxide plasma concentrations increase in combination with drugs that inhibit CYP enzymes, including cimetidine, disulfiram, isoniazid, estrogen, and oral contraceptives.15

Nefazodone—a CYP 3A3/4 inhibitor—can increase plasma concentrations of triazolam and alprazolam to potentially toxic levels. Nefazodone’s manufacturer recommends lowering triazolam dosages by 75% and alprazolam dosages by 50% when used with nefazodone.3

Carbamazepine—a CYP 3A3/4 inducer—induces both its own and other drugs’ metabolism. It can lower plasma concentrations of alprazolam, clonazepam, midazolam, and triazolam, which are metabolized by 3A3/4. Smoking, food, and antacids also may decrease benzodiazepine plasma concentrations.

As perpetuator drugs, benzodiazepines might increase digoxin plasma concentration, probably because of reduced digoxin renal clearance.16 Diazepam may inhibit CYP 2C9 and/or 2C19 by being an alternate substrate for enzymebinding sites,15,17 increasing the concentration of other drugs such as phenytoin.

Buspirone: Complicated pharmacology

One of buspirone’s major clinical advantages is that it does not pharmacodynamically or pharmacokinetically affect benzodiazepines. Buspirone, the only azaspirodecanedione marketed in the United States, has complex central 5-HT effects.18,19 Because it is a partial 5-HT1A agonist, buspirone’s net effect depends on 5-HT concentration at the receptor:

  • If 5-HT concentration is low, buspirone will act as an agonist.
  • If 5-HT concentration is high, buspirone—being a partial agonist—will antagonize the effect of excessive 5-HT.
Buspirone also acts at postsynaptic and presynaptic 5-HT1A receptors, which mediate different physiologic mechanisms in the brain. Finally, buspirone may act more as a full agonist at postsynaptic than at presynaptic 5-HT1A receptors.20

Buspirone’s pharmacology is further complicated by its conversion via oxidative metabolism into an active metabolite—1-pheyl-piperazine (1-PP). Buspirone is a CYP 3A3/4 enzyme substrate, so it is extensively metabolized as it crosses the duodenum and passes through the liver. As a result, the parent drug has low bioavailability and is principally converted into 1-PP before entering systemic circulation.6

1-PP works differently than the parent drug. As an alpha-2-adrenergic antagonist, 1-PP increases the firing rate of adrenergic neurons in the locus ceruleus by blocking a receptor in presynaptic feedback system.

Which traits of buspirone and its active metabolite produce the drug’s anxiolytic effect? It might be one of these, all of them, or some other unknown trait.

Pharmacodynamic DDIs. Presumably because of its effects on serotonin release at 5-HT1A receptors, buspirone may cause hypertensive episodes when used with monoamine oxidase inhibitors (MAOIs) (Table 3). This is why a 2-week washout is recommended between discontinuing an MAOIs and starting buspirone.21

Table 3

Clinical effects of drug-drug interactions with buspirone

Pharmacodynamic
DO NOT use buspirone with monoamine oxidase inhibitors (MAOIs); allow 2-week washout after stopping an MAOI before starting buspirone
Pharmacokinetic
Food ↑ buspirone Cmax and AUC 2-fold
Renal impairment ↑ buspirone plasma concentration 2-fold
Hepatic impairment ↑ buspirone Cmax and AUC 15-fold and ↑ half-life 2-fold
Verapamil, diltiazem, erythromycin, itraconazole ↑ buspirone plasma concentration
Rifampicin ↓ buspirone plasma concentration 10-fold
Buspirone ↑ haloperidol plasma concentration
Erythromycin, itraconazole, nefazodone, grapefruit juice ↑ buspirone plasma concentration
Cmax: maximum drug concentration
AUC: area under the curve (mathematical calculation of the body’s total exposure to a drug over time)
In theory, buspirone might cause serotonin syndrome when combined with MAOIs. Rare cases of serotonin syndrome have been reported in patients taking buspirone and selective serotonin reuptake inhibitors (SSRIs) and/or trazodone.6 On the other hand, using buspirone to augment SSRIs can cause therapeutic DDIs. Some researchers have added buspirone when patients have not benefited from SSRI monotherapy because:

  • buspirone affects 5-HT mechanisms
  • drugs that affect serotonin reuptake inhibition, 5HT1A receptors, and 5HT2 receptors may have synergy.20
Pharmacokinetic DDIs. Avoid combining buspirone with verapamil, diltiazem, erythromycin, or itraconazole because competitive enzyme inhibition will substantially increase buspirone’s plasma concentration.21

Some SSRIs—such as high-dose fluoxetine and usual doses of fluvoxamine—may increase buspirone serum concentration by inhibiting CYP 3A4.6 Consider this clinical effect before you combine an SSRI with buspirone. Using buspirone with fluoxetine, paroxetine, or bupropion also increases serum 1-PP. This increase, which occurs when CYP 2D6 slows 1-PP clearance, could cause euphoria, mania, or seizures.20

Coadministering rifampin can lower buspirone plasma concentrations almost 10-fold because rifampin induces CYP 3A3/4.22

As a perpetuator, buspirone can increase haloperidol plasma concentrations, but probably not to a clinically important extent. In an open trial, Goff23 added buspirone, mean dosage 23.8 mg/d, to a stable regimen of haloperidol in 7 patients with schizophrenia. Although haloperidol’s mean plasma concentration increased by 26% after 6 weeks, this modest change would be difficult to detect in clinical practice.

 

 

Huang et al24 found no clinically significant pharmacokinetic interaction between buspirone, 10 mg tid, and haloperidol, 10 to 40 mg/d, during 6 weeks of coadministration in 27 patients with schizophrenia.

Propranolol: Beta-blocking anxiolytic

Propranolol is prescribed off-label for anxiety disorders more often than other beta blockers. It may help patients with situational or performance anxiety.

Beta-adrenergic blockers competitively antagonize norepinephrine and epinephrine at the beta-adrenergic receptor. These cardiovascular agents can reduce many of anxiety’s peripheral manifestations, such as tachycardia, diaphoresis, trembling, and blushing. All beta blockers share this pharmacologic effect, but their pharmacokinetics differ greatly. Some depend on a single CYP enzyme for clearance (metoprolol, by CYP 2D6), whereas others, such as propranolol, are metabolized by multiple CYP enzymes.

Pharmacodynamic DDIs. Drugs that block alpha-1 adrenergic receptors potentiate beta blockers’ blood pressure-lowering effects and increase the risk of orthostatic hypotension. This is probably why haloperidol can potentiate propranolol’s hypotensive effects.6 Other alpha-1 adrenergic antagonists—though not normally classified as such—include some tertiary amine tricyclic antidepressants (amitriptyline and imipramine) and some antipsychotics (quetiapine).

Reports have associated hypertensive crises and bradycardia with coadministration of beta blockers and MAOIs.21 Depressed myocardial contractility and A-V nodal conduction may occur when beta blockers are combined with calcium channel inhibitors.21 Beta blockers also can decrease IV anesthetic dose requirements because they decrease cardiac output.25

In patients using insulin for diabetes mellitus, propranolol inhibits recovery from insulin-induced hypoglycemia and may cause hypertension and bradycardia. Beta blockers also can mask the tachycardia that usually accompanies insulin-induced hypoglycemia.

Pharmacokinetic DDIs. Propranolol has an extensive first-pass effect, being etabolized in the liver to active and inactive compounds that interact with CYP enzymes 1A2, 2C18, 2C19 and 2D6.6

Coadministering strong CYP 2D6 inhibitors such as bupropion, fluoxetine, or paroxetine can reduce propanolol clearance, increasing its effect and risking cardiac toxicity6 (Table 4). CYP 1A2 inhibitors such as amiodarone and fluoroquinolones or CYP 2C19 inhibitors such as fluvoxamine also increase serum concentrations of propranolol.

Table 4

How to avoid drug interactions with three common anxiolytics*

When prescribing benzodiazepines…
DODO NOT
Advise patients not to combine benzodiazepines with alcoholUse with other CNS depressants or nefazodone
Talk to patients about potential for abuse/dependency, and monitor benzodiazepine useUse in elderly patients or in patients with high potential for substance abuse
When prescribing buspirone…
DODO NOT
Allow a 2-week washout between discontinuing an MAOI and starting buspironeUse with MAOIs, verapamil, diltiazem, erythromycin, or itraconazole
Consider adding buspirone when SSRI monotherapy has not adequately helped patients with anxietyCo-administer with rifampin
Combine with benzodiazepines, if needed 
When prescribing propranolol…
DODO NOT
Educate patients using insulin for diabetes mellitus that propranolol may inhibit recovery from insulin-induced hypoglycemia, cause bradycardia, or mask tachycardiaCombine with medications with strong hypotensive effects
 Coadminister with strong CYP 2D6 or 1A2 inhibitors
Recheck anticonvulsant plasma concentrations after starting propranololAdd to calcium inhibitors for patients with ↓ myocardial contractility and A-V nodal conduction
* Before prescribing any anxiolytic, review all co-prescribed medications for potential DDIs
DDI: drug-drug interaction
MAOI: monoamine oxidase inhibitor
SSRI: selective serotonin reuptake inhibitor
On the other hand, CYP inducers such as barbiturates, phenytoin, and cigarette smoking can increase propranolol elimination and decrease its serum levels.26 Hyperthyroidism may enhance propranolol’s presystemic clearance but has little effect on its half life.27

As a perpetuator, propranolol produces small increases in diazepam concentration, suggesting that the beta-blocker inhibits diazepam metabolism. This interaction can impair kinetic visual acuity, which is correlated with the ability to discriminate moving objects in space.26

Propranolol increases plasma concentrations of antipsychotics, anticonvulsants, theophylline, and levothyroxine (Table 5)—possibly because of the beta blocker’s negative inotropic effects (decreased cardiac output reduces hepatic and renal blood flow).

Table 5

Clinical effects of drug-drug interactions with propranolol

Pharmacodynamic
With MAO inhibitors → hypertensive crisis and bradycardia
With calcium channel inhibitors → ↓ myocardial contractility and A-V nodal conduction
↓ intravenous anesthetic dose requirements
↓ diazepam metabolism
↓ median effective dosage of valproate and diazepam; might improve antiepileptic potential of valproate
Pharmacokinetic
↑ plasma concentration of antipsychotics, anticonvulsants, theophylline, levothyroxine
Barbiturates, phenytoin, and cigarette smoking ↑ propranolol elimination
Related resources

Drug brand names

  • Alprazolam • Xanax
  • Bupropion • Wellbutrin
  • Buspirone • BuSpar
  • Carbamazepine • Carbatrol, others
  • Chlordiazepoxide • Librium
  • Cimetidine • Tagamet
  • Clonazepam • Klonopin
  • Clorazepate • Tranxene
  • Clozapine • Clozaril
  • Diazepam • Valium
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Itraconazole • Sporanox
  • Lorazepam • Ativan
  • Midazolam • Versed
  • Mirtazapine • Remeron
  • Oxazepam • Serax
  • Paroxetine • Paxil
  • Phenytoin • Dilantin
  • Propranolol • Inderal
  • Quetiapine • Seroquel
  • Rifampin • Rifadin, Rimactane
  • Triazolam • Halcion
  • Valproate • various
  • Verapamil • Calan, Isoptin
Disclosures

Drs. Ramadan and Werder report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

 

 

Dr. Preskorn has received grants or has been a consultant or speaker for Abbott Laboratories, AstraZeneca Pharmaceuticals, Boehringer-Ingelheim, Bristol-Myers Squibb Co., Merck & Co., Eisai, Eli Lilly and Co., GlaxoSmithKline, Janssen Pharmaceutica, Johnson & Johnson, Novartis Pharmaceuticals Corp., Organon, Otsuka America Pharmaceutical, Pfizer, Solvay Pharmaceuticals, Sanofi-Aventis, and Wyeth.

References

1. Preskorn S, Flockhart D. Psychiatric drug interactions guide. New York: MBL Communications.; 2004.

2. Bruce SE, Yonkers KA, Otto MW, et al. Influence of psychiatric comorbidity on recovery and recurrence in generalized anxiety disorder, social phobia, and panic disorder 12-year prospective study. Am J Psychiatry 2005;162:1179-87.

3. Nemeroff CB. Use of atypical antipsychotics in refractory depression and anxiety. J Clin Psychiatry 2005;66(Suppl 8):13-21.

4. Bean RB, Bean WB. Sir William Osler: Aphorisms from his bedside teaching and writing. Springfield, IL: Charles C. Thomas; 1961:53.

5. Tasman A, Kay J, Lieberman JA. Psychiatry Therapeutics. 2nd ed. West Sussex, UK: John Wiley & Sons; 2003:347.

6. Fuller M, Sajatovic M. Drug information for mental health. 3rd ed. Hudson, OH: Lexi-Comp; 2001.

7. Stahl SM. Essential psychopharmacology: Neuroscientific basis and practical applications. 2nd ed. New York: Cambridge University Press; 2000.

8. Janicak PG, Davis JM, Preskorn SH, Ayad FJ. Principles and practice of psychopharmacology. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2001.

9. Tanaka E. Toxicological interactions involving psychiatric drugs and alcohol: an update. J Clin Pharm Ther 2003;28(2):81-95.

10. Frels C, Williams P, Narayanan S, Gariballa SE. Iatrogenic causes of falls in hospitalised elderly patients: a case-control study. Postgrad Med 2002;78(922):487-9.

11. Wolf BC, Lavezzi WA, Sullivan LM, Flannagan LM. One hundred seventy two deaths involving the use of oxycodone in Palm Beach County. J Forensic Sci 2005;50(1):192-5.

12. Rogers WO, Hall MA, Brissie RM, Robinson CA. Detection of alprazolam in three cases of methadone/benzodiazepine overdose. J Forensic Sci 1997;42(1):155-6.

13. Wolf BC, Lavezzi WA, Sullivan LM, et al. Alprazolam-related deaths in Palm Beach County. Am J Forensic Med Pathol 2005;26(1):24-7.

14. Isbister GK, O’Regan L, Sibbritt D, et al. Alprazolam is relatively more toxic than other benzodiazepines in overdose. Br J Clin Pharmacol 2004;58(1):88-95.

15. Sadock BJ, Sadock VA. Kaplan and Sadock’s pocket handbook of psychiatric drug treatment. 3rd ed. Philadelphia: Lippincott Williams & Wilkins; 2001.

16. Tollefson G, Lesar T, Grothe D, et al. Alprazolam-related digoxin toxicity. Am J Psychiatry 1984;141(12):1612-3.

17. Murphy A, Wilbur K. Phenytoin-diazepam interaction. Ann Pharmacother 2003;37(5):659-3.

18. Sharp T, McQuade R, Bramwell S, et al. Effect of acute and repeated administration of 5-HT1A receptor agonists on 5-HT release in rat brain in vivo. Naunhyn Schmiedebergs Arch Pharmacol 1993;348(4):339-46.

19. Van den Hooff P, Galvan M. Actions of 5-hydroxytryptamine and 5-HT1A receptor ligands on rat dorso-lateral septal neurons in vitro. Br J Pharmacol 1992;106(4):893-9.

20. Preskorn SH Do you believe in magic? Journal of Practical Psychiatry and Behavioral Health March 1997;99-103

21. Physicians’ Desk Reference. 59th ed. Montvale, NJ: Thomson PDR; 2005.

22. Mahmood I, Sahajwalla C. Clinical pharmacokinetics and pharmacodynamics of buspirone, an anxiolytic drug. Clin Pharmacokinet 1999;36(4):277-87.

23. Goff DC, Midha KK, Brotman AW, et al. An open trial of buspirone added to neuroleptics in schizophrenic patients. J Clin Psychopharmacol 1991;11(3):193-7.

24. Huang HF, Jann MW, Wei FC, et al. Lack of pharmacokinetic interaction between buspirone and haloperidol in patients with schizophrenia. J Clin Pharmacol 1996;36(10):963-9.

25. Avram MJ, Krejcie TC, Henthorn TK, et al. Beta-adrenergic blockade affects initial drug distribution due to decreased cardiac output and altered blood flow distribution. J Pharmacol Exp Ther 2004;311(2):617-24.

26. Hawksworth G, Betts T, Crowe A, et al. Diazepam/beta-adrenoceptor antagonist interactions. Br J Clin Pharmacol 1984;17(Suppl 1):69S-76S.

27. Hallengren B, Nilsson OR, Karlberg BE, et al. Influence of hyperthyroidism on the kinetics of methimazole, propranolol, metoprolol and atenolol. Eur J Clin Pharmacol 1982;21(5):379-84.

References

1. Preskorn S, Flockhart D. Psychiatric drug interactions guide. New York: MBL Communications.; 2004.

2. Bruce SE, Yonkers KA, Otto MW, et al. Influence of psychiatric comorbidity on recovery and recurrence in generalized anxiety disorder, social phobia, and panic disorder 12-year prospective study. Am J Psychiatry 2005;162:1179-87.

3. Nemeroff CB. Use of atypical antipsychotics in refractory depression and anxiety. J Clin Psychiatry 2005;66(Suppl 8):13-21.

4. Bean RB, Bean WB. Sir William Osler: Aphorisms from his bedside teaching and writing. Springfield, IL: Charles C. Thomas; 1961:53.

5. Tasman A, Kay J, Lieberman JA. Psychiatry Therapeutics. 2nd ed. West Sussex, UK: John Wiley & Sons; 2003:347.

6. Fuller M, Sajatovic M. Drug information for mental health. 3rd ed. Hudson, OH: Lexi-Comp; 2001.

7. Stahl SM. Essential psychopharmacology: Neuroscientific basis and practical applications. 2nd ed. New York: Cambridge University Press; 2000.

8. Janicak PG, Davis JM, Preskorn SH, Ayad FJ. Principles and practice of psychopharmacology. 3rd ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2001.

9. Tanaka E. Toxicological interactions involving psychiatric drugs and alcohol: an update. J Clin Pharm Ther 2003;28(2):81-95.

10. Frels C, Williams P, Narayanan S, Gariballa SE. Iatrogenic causes of falls in hospitalised elderly patients: a case-control study. Postgrad Med 2002;78(922):487-9.

11. Wolf BC, Lavezzi WA, Sullivan LM, Flannagan LM. One hundred seventy two deaths involving the use of oxycodone in Palm Beach County. J Forensic Sci 2005;50(1):192-5.

12. Rogers WO, Hall MA, Brissie RM, Robinson CA. Detection of alprazolam in three cases of methadone/benzodiazepine overdose. J Forensic Sci 1997;42(1):155-6.

13. Wolf BC, Lavezzi WA, Sullivan LM, et al. Alprazolam-related deaths in Palm Beach County. Am J Forensic Med Pathol 2005;26(1):24-7.

14. Isbister GK, O’Regan L, Sibbritt D, et al. Alprazolam is relatively more toxic than other benzodiazepines in overdose. Br J Clin Pharmacol 2004;58(1):88-95.

15. Sadock BJ, Sadock VA. Kaplan and Sadock’s pocket handbook of psychiatric drug treatment. 3rd ed. Philadelphia: Lippincott Williams & Wilkins; 2001.

16. Tollefson G, Lesar T, Grothe D, et al. Alprazolam-related digoxin toxicity. Am J Psychiatry 1984;141(12):1612-3.

17. Murphy A, Wilbur K. Phenytoin-diazepam interaction. Ann Pharmacother 2003;37(5):659-3.

18. Sharp T, McQuade R, Bramwell S, et al. Effect of acute and repeated administration of 5-HT1A receptor agonists on 5-HT release in rat brain in vivo. Naunhyn Schmiedebergs Arch Pharmacol 1993;348(4):339-46.

19. Van den Hooff P, Galvan M. Actions of 5-hydroxytryptamine and 5-HT1A receptor ligands on rat dorso-lateral septal neurons in vitro. Br J Pharmacol 1992;106(4):893-9.

20. Preskorn SH Do you believe in magic? Journal of Practical Psychiatry and Behavioral Health March 1997;99-103

21. Physicians’ Desk Reference. 59th ed. Montvale, NJ: Thomson PDR; 2005.

22. Mahmood I, Sahajwalla C. Clinical pharmacokinetics and pharmacodynamics of buspirone, an anxiolytic drug. Clin Pharmacokinet 1999;36(4):277-87.

23. Goff DC, Midha KK, Brotman AW, et al. An open trial of buspirone added to neuroleptics in schizophrenic patients. J Clin Psychopharmacol 1991;11(3):193-7.

24. Huang HF, Jann MW, Wei FC, et al. Lack of pharmacokinetic interaction between buspirone and haloperidol in patients with schizophrenia. J Clin Pharmacol 1996;36(10):963-9.

25. Avram MJ, Krejcie TC, Henthorn TK, et al. Beta-adrenergic blockade affects initial drug distribution due to decreased cardiac output and altered blood flow distribution. J Pharmacol Exp Ther 2004;311(2):617-24.

26. Hawksworth G, Betts T, Crowe A, et al. Diazepam/beta-adrenoceptor antagonist interactions. Br J Clin Pharmacol 1984;17(Suppl 1):69S-76S.

27. Hallengren B, Nilsson OR, Karlberg BE, et al. Influence of hyperthyroidism on the kinetics of methimazole, propranolol, metoprolol and atenolol. Eur J Clin Pharmacol 1982;21(5):379-84.

Issue
Current Psychiatry - 05(05)
Issue
Current Psychiatry - 05(05)
Page Number
16-28
Page Number
16-28
Publications
Publications
Article Type
Display Headline
Protect against drug-drug interactions with anxiolytics
Display Headline
Protect against drug-drug interactions with anxiolytics
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Drug-drug interactions: Avoid serious adverse events with mood stabilizers

Article Type
Changed
Tue, 12/11/2018 - 15:10
Display Headline
Drug-drug interactions: Avoid serious adverse events with mood stabilizers

Drug-drug interactions (DDIs) can be viewed as physiologic combat wherein a “perpetrator” drug affects a “victim” drug’s pharmacokinetics or pharmacodynamics. Your challenge is to deter that interaction in patients taking two or more medications.

This article—first in a series—discusses polypharmacy risk factors that increase the likelihood of detrimental DDIs, then focuses on DDIs in patients taking mood stabilizers for bipolar disorder. We also offer practical tips to reduce DDI risk. Future articles will discuss DDI risks with antidepressants, antipsychotics, and anxiolytics.

To predict DDIs, you need to know psychotropics’ mechanism of action, metabolism, and effects on cytochrome P-450 (CYP) enzymes. Our discussion is not exhaustive because the data base is massive and new interactions continue to be discovered. Our aim is to equip you to anticipate and prevent DDIs when prescribing.

WHAT ARE ADVERSE DDIs?

An adverse event (AE) is any undesirable experience that occurs when a patient uses a medical product, whether or not the product caused the event. The FDA says an “undesirable experience” may be:

  • an unfavorable and unintended symptom or sign
  • an abnormal lab or radiographic finding
  • a disease that is temporarily associated with the medical product.
A temporal relationship is all that is required, although preexisting conditions and events clearly related to other causes are not usually considered adverse events.

An AE becomes “serious” (an SAE) when its duration, intensity, and/or frequency leads to death, a life-threatening condition, initial or prolonged hospitalization, disability, or congenital anomaly. Reporting is voluntary, but we strongly recommend that you report all SAEs to the FDA.

These definitions can help you confirm that a patient has experienced an SAE, but the task becomes more complicated when you try to attribute an SAE to a drug interaction. In the absence of an FDA definition, we assert that DDIs are responsible for SAEs when a perpetrator drug affects the pharmacokinetics or pharmacodynamics of a victim drug and exacerbates a known untoward event of the victim drug (Box 1).1-5 Which drug is the perpetrator and which is the victim is not always clear, and sometimes a medication—such as carbamazepine—can be both at once.

Box 1

Drug-drug interactions: Taking a toll

More than 100,000 possible detrimental DDIs have been documented in medical literature and pharmaceutical company data. This number is likely to grow with increased scrutiny, as

DDIs cause morbidity, mortality, and increased health care costs. More than 106,000 Americans die each year from properly prescribed, correctly taken medications. Polypharmacy is associated with extended hospital stays, and using >6 drugs is an independent predictor of death. DDIs contribute to the cause of death in acute overdoses and can be responsible for false-positive suicide diagnoses.

In clinical practice, DDI-associated toxicity may be mistaken for a new disease process, or a disease may be incorrectly perceived as progressing when a medication is rendered ineffective.

Source: References 1-5

RISKS OF POLYPHARMACY

Individuals with psychiatric illnesses are at particular risk for DDIs (Box 2). Patients seen by psychiatrists, for example, are six times more likely than patients seen by primary care physicians to be taking multiple medications.6

Polypharmacy increases the risk of adverse events, nonadherence, medication errors, and drug interactions.7 FDA’s MedWatch Web site lists more than 630 DDI warnings.8 The more medications a patient is taking, the greater the risk for detrimental DDIs and cumulative toxicity,9 which often lead to DDI-induced AEs.10

A study of DDIs in 5,125 mostly older outpatients11 found that:

  • 1,594 (31%) had at least one interacting drug combination (average 1.6)
  • subjects with one or more DDIs were taking an average 8.1 drugs, compared with 5.2 drugs in those without DDIs—a significant difference
  • 155 (3%) had interactions of “major clinical significance.”
‘Uncontrolled experiments.’ Drug combinations often are “uncontrolled experiments” with unknown potential for toxic effects.12 Studies have linked polypharmacy and DDIs as well as DDIs and AEs:

  • Although drug interactions are responsible for only 3.8% of emergency department visits, patients with DDIs are usually admitted to the hospital.13
  • Preventable drug interactions cause approximately one-third of all AEs in hospitalized patients and account for one-half of all AE costs.14
DDI risk is increasing over time as the number of medications used to treat psychiatric patients has grown. For example, 3.3% of patients discharged between 1974 and 1979 from the National Institute of Mental Health Biological Psychiatry Branch were taking 3 or more medications, compared with more than 40% of patients discharged between 1990 and 1995—a 12-fold increase.15

Box 2

Psychiatric patients: High risk for DDIs

Symptom-based prescribing. Patients with psychiatric illness are often prescribed >1 medication to manage symptoms and signs, rather than a single medication targeting a specific psychiatric disorder.

Multiple prescribers. Patients with anxiety and depressive disorders may see multiple providers, which increases the risk for polypharmacy, drug-drug interactions, and adverse events.

Medical comorbidity. Persons with psychiatric illness are at increased risk for concomitant medical illness, and persons with medical illness are at increased risk for psychiatric illness.

Psychiatric comorbidity. Persons with one psychiatric illness are at increased risk for other psychiatric illnesses.

Source: Adapted from reference 6.

 

 

HOW TO MINIMIZE DDI RISK

Use the acronym “LISTEN” (Table 1) to minimize DDI risk in patients taking combination therapies.16 The 6 steps in LISTEN can help you determine which drug or drugs you may discontinue before adding another.

We also recommend that you monitor therapeutic and toxic effects by checking serum drug levels, especially for drugs with a low therapeutic index. Lithium, for example, requires close mentoring of plasma concentration every 2 to 6 months and during dosage adjustments to avoid toxicity.17 Therapeutic drug monitoring has been shown to prevent adverse events from DDIs.16 For added safety, encourage patents to purchase all medications at one pharmacy and to enroll in that pharmacy’s DDI monitoring program.18

Keep in mind that systemic conditions may require a dosage change:

  • Increased volume of distribution, as in patients who gain weight or total water volume, requires higher doses to maintain a constant therapeutic effect.
  • Reduced clearance, as in patients with decreased renal or hepatic function, will likely require lower doses to prevent toxicity.19
Table 1

LISTEN: 6 tips to minimize DDI risk

LListeach drug’s name and dosage in the patient’s chart and in a note given to the patient.
IEach drug should have a clear indication and well-defined therapeutic goal; discontinue any drug not achieving its goal
SMake the regimen as simple as possible, with once- or twice-daily dosing.
TWhen possible, treat multiple symptoms with a single drug, rather than multiple symptoms with multiple drugs
EEducatepatients about polypharmacy, DDIs, and adverse events; assess all medications—including vitamins, minerals, herbs, dietary supplements, nonprescription products—and address potential DDIs
NAvoid prescribing medications with a narrow therapeutic window.

DDIsWITH MOOD STABILIZERS

Diagnoses of schizophrenia, anxiety disorders, and affective disorders are major risk factors for polypharmacy.20 DDIs are a particular concern in patients with bipolar disorder, given their complex treatment regimens.21

Interactions occur with the most commonly prescribed bipolar medications, including lithium and anticonvulsants (Table 2).17.21-25 Although atypical antipsychotics are also considered mood stabilizers in bipolar disorder, we will discuss their potential DDIs in a future article.

Table 2

Some drug-drug interactions with mood stabilizers

Mood stabilizerDrug interactions
Carbamazepine↑plasma clomipramine, phenytoin, primidone
↑risk of neurotoxic side effects and confusional states with lithium
Alters thyroid function with anticonvulsants
↓anticoagulant concentrations and↑bleeding risk
↓oral contraceptive reliability; can cause false-negative pregnancy tests
↑metabolism and may ↓efficacy of cancer chemotherapy (docetaxel, estrogens, paclitaxel, progesterone, cyclophosphamide)
↑aprepitant, granisetron metabolism and ↓efficacy
↑glipizide, tolbutamide metabolism
LithiumNSAIDs (ibuprofen, indomethacin, piroxicam) and COX-2 inhibitors ↑plasma lithium
ACE inhibitors ↑plasma lithium
Calcium channel blockers and carbamazepine ↑lithium neurotoxicity
SSRIs ↑diarrhea, confusion, tremor, dizziness, and agitation
Acetazolamide, urea, xanthine preparations, alkalinizing agents such as sodium bicarbonate ↓plasma lithium
Metronidazole ↑lithium toxicity
Encephalopathic syndrome possible with haloperidol
Lamotrigine↑concentration of carbamazepine’s epoxide metabolite
Carbamazepine, phenytoin, phenobarbital ↓plasma lamotrigine 40% to 50%
↑plasma sertraline
↓plasma valproic acid 25%; valproic acid doubles plasma lamotrigine and ↑rash risk
Topiramate↑valproic acid concentrations 11%; valproic acid ↓plasma topiramate 14%
↑plasma phenytoin up to 25%; phenytoin, carbamazepine ↓plasma topiramate by 40% to 48%
↓digoxin bioavailability
↓oral contraceptive efficacy
Valproic acid↑plasma phenobarbital, primidone
↓phenytoin clearance, volume distribution and ↑breakthrough seizure risk
↑serum concentration of antiepileptics, such as lamotrigine; absence status possible with clonazepam
↑= Increases ↓= Decreases
ACE = angiotensin-converting enzyme; COX = cyclooxygenase
NSAIDs = nonsteroidal anti-inflammatory drugs; SSRIs = selective serotonin reuptake inhibitors
Source: References 17, 21-25

LITHIUM: TOXICITY RISK

Lithium is excreted via the kidneys, so be cautious when using lithium in patients taking diuretics.17,22 Drugs that can lower serum lithium concentrations by increasing urinary lithium excretion include acetazolamide, urea, xanthine preparations, and alkalinizing agents such as sodium bicarbonate.17

Combining lithium with selective serotonin reuptake inhibitors can cause diarrhea, confusion, tremor, dizziness, and agitation.17 An encephalopathic syndrome has occurred in a few patients treated with lithium plus haloperidol.

Monitor lithium levels closely when bipolar patients start or stop nonsteroidal anti-inflammatory drugs (NSAIDs). Nonprescription ibuprofen can cause serious and even life-threatening serum lithium elevations by affecting lithium’s rate of tubular reabsorption.26 Indomethacin, piroxicam, and selective cyclooxygenase-2 (COX-2) inhibitors also increase plasma lithium concentrations.25

For patients taking lithium with heart drugs, angiotensin-converting enzyme (ACE) inhibitors may increase plasma lithium levels,17 and calcium channel blockers may increase the neurotoxicity risk.17,22 Using the anti-infective metronidazole with lithium may provoke lithium toxicity.

VALPROIC ACID: MONITOR CLEARANCE

Drugs that affect the expression of hepatic enzymes—especially glucuronosyltransferase—may increase clearance of valproic acid and its derivatives. Phenytoin, carbamazepine, or phenobarbital, for example, can double valproic acid clearance.

On the other hand, drugs that inhibit CYP-450 (such as antidepressants) have little effect on valproic acid concentration. Valproate can decrease plasma clearance of amitriptyline, so consider monitoring this tricyclic’s blood levels in patients also taking valproate.17

Because valproic acid can increase serum phenobarbital, monitor barbiturate concentrations when using these two drugs. A similar interaction occurs with primidone, which is metabolized into a barbiturate. Breakthrough seizures may occur with phenytoin, as valproic acid can reduce phenytoin clearance and apparent volume distribution by 25%.22

 

 

Using valproic acid with clonazepam may produce absence status in patients with a history of absence-type seizures.17 Valproic acid also displaces diazepam from its plasma albumin binding sites and inhibits its metabolism.

Concomitant use of valproic acid can increase serum concentrations of other antiepileptic drugs. For example, lamotrigine levels may double,24 and felbamate’s peak concentration may increase and require dosage reduction. Valproic acid may also interact with nonpsychiatric medications:

  • Subtherapeutic valproic acid levels have been reported when co-administered with the antibiotic meropenem.
  • In patients with HIV infection, valproic acid can decrease clearance of the antiretroviral zidovudine by 38%.
  • Patients receiving rifampin for tuberculosis may need a dosage adjustment, as oral rifampin’s clearance can increase 40% with concomitant valproic acid.

CARBAMAZEPINE: SELF-INDUCER

Metabolized by CYP 3A4, carbamazepine may induce its own metabolism as well as the CYP 3A4 isoenzyme. Therefore inhibitors and inducers of CYP 3A4 may affect carbamazepine plasma levels.

Carbamazepine can increase plasma levels of other psychotropics including clomipramine, phenytoin, and primidone.17,22 When used with lithium, it may increase the risk of neurotoxic side effects and confusion.23 It can alter thyroid function when used with other anticonvulsants.

For bipolar patients with diabetes, carbamazepine can cause hyperglycemia by inducing the metabolism of oral sulfonylureas such as glipizide and tolbutamide. In women, carbamazepine decreases the reliability of oral contraceptives17 and can cause false-negative pregnancy tests.24

For cancer patients, concurrent carbamazepine may induce metabolism of chemotherapy drugs such as docetaxel, estrogens, paclitaxel, progesterone, and cyclophosphamide, decreasing their efficacy.21 It can increase metabolism of aprepitant and granisetron—used to treat chemotherapy-related nausea—reducing plasma concentrations and possibly efficacy. Carbamazepine’s additive dopamine blockade can increase the risk of extrapyramidal symptoms when used with docetaxel or the antiemetic/antivertigo agents chlorpromazine, metoclopramide, or prochlorperazine.

Carbamazepine increases elimination of some cardiovascular drugs and may decrease the effect of antiarrhythmics such as lidocaine and quinidine; calcium channel blockers such as amlodipine, nifedipine, felodipine, nisoldipine, diltiazem, and verapamil; the beta blocker propranolol; and the vasodilator bosetan.21 Carbamazepine also reduces anticoagulant concentrations, and breakthrough bleeding has been reported.

(See "Out of the Pipeline−extended−release carbamazepine" for a listing of drugs that interact with this agent.)

OTHER ANTICONVULSANTS

Lamotrigine. Some concomitant CNS medications—such as carbamazepine, phenytoin or phenobarbital—reduce lamotrigine serum concentrations by as much as 50%.17 This substantial reduction may give the impression that the patient is not responding to therapeutic lamotrigine doses.

Patients taking lamotrigine with carbamazepine may be at greater risk for dizziness, diplopia, ataxia, and blurred vision because of increased serum concentration of carbamazepine’s epoxide metabolite. Valproic acid doubles lamotrigine serum concentration and increases the risk of rash, whereas lamotrigine decreases valproic acid concentration by 25%.17 Lamotrigine’s manufacturer offers special starting kits for patients taking carbamazepine or valproic acid.

Sertraline increases plasma lamotrigine concentration—but to a lesser extent than does valproic acid17 —and no dosage adjustment is needed.

Topiramate. Concomitant carbamazepine or phenytoin reduces topiramate concentration by 40% to 48%, whereas topiramate increases phenytoin concentration up to 25%. Similarly, valproic acid reduces topiramate’s concentration by 14%, while at the same time valproic acid concentration increases by 11%.17

Topiramate slightly decreases digoxin’s bioavailability and the efficacy of estrogenic oral contraceptives.17,22

Related resources

Drug brand names

  • Aprepitant • Emend
  • Bosentan • Tracleer
  • Carbamazepine • Tegretol, others
  • Chlorpromazine • Thorazine
  • Clomipramine • Anafranil, others
  • Clonazepam • Klonopin
  • Cyclophosphamide • Cytoxan, Neosar
  • Diazepam • Valium
  • Diltiazem • Cardizem, others
  • Docetaxel • Taxotere
  • Felbamate • Felbatol
  • Felodipine • Plendil
  • Granisetron • Kytril
  • Glipizide • Glucotrol
  • Haloperidol • Haldol
  • Indomethacin • Indocin
  • Lamotrigine • Lamictal
  • Meropenem • Merrem
  • Metoclopramide • Reglan
  • Metronidazole • Flagyl
  • Nifedipine • Adalat, Procardia
  • Nisoldipine • Sular
  • Paclitaxel • Taxol, others
  • Phenobarbital • Solfoton
  • Phenytoin • Dilantin
  • Piroxicam • Feldene
  • Primidone • Mysoline
  • Prochlorperazine • Compazine
  • Propranolol • Inderal
  • Rifampin • Rifadin
  • Sertraline • Zoloft
  • Tolbutamide • Orinase
  • Topiramate • Topamax
  • Valproic acid • Depakote
  • Verapamil • Calan, others
  • Zidovudine • Retrovir
Disclosure

Dr. Ramadan and Dr. Werder report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Preskorn has received grants or has been a consultant or speaker for Abbott Laboratories, AstraZeneca, Boehringer-Ingelheim, Bristol-Myers Squibb Co., Merck & Co., Eisai Inc., Eli Lilly and Co., GlaxoSmithKline, Janssen Pharmaceutica, Johnson & Johnson, Novartis Pharmaceuticals, Organon, Otsuka America Pharmaceutical Inc., Pfizer Inc., Solvay Pharmaceuticals, Sanofi-Aventis, and Wyeth.

References

1. Langdorf MI, Fox JC, Marwah RS, et al. Physician versus computer knowledge of potential drug interactions in the emergency department. Acad Emerg Med 2000;7:1321-9.

2. Lazarou J, Pomeranz BH, Cory PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 1998;279(15):1200-5.

3. Incalzi RA, Gemma A, Capparella O, et al. Predicting mortality and length of stay of geriatric patients in an acute care general hospital. J Gerontol 1992;47(2):M35-9.

4. Preskorn SH. Fatal drug-drug interaction as a differential consideration in apparent suicide. J Psychiatr Pract 2002;8(4):233-8.

5. Peterson JF, Bates D. Preventable medication errors: identifying and eliminating serious drug interactions. J Am Pharm Assoc (Wash) 2001;41(2):159-60.

6. Nichol MB, Stimmel GL, Lange SC. Factors predicting the use of multiple psychotropic medications. J Clin Psychiatry 1995;2:60-6.

7. Ananth J, Parameswaran S, Gunatilake S. Antipsychotic polypharmacy. Curr Pharm Des 2004;10(18):2231-8.

8. Medwatch Web site. Food and Drug Administration. Search for Drug-drug interactions. Available at: http://www.fda.gov/med-watch/index.html. Accessed March 31, 2005.

9. Rascati K. Drug utilization review of concomitant use of specific serotonin reuptake inhibitors or clomipramine and antianxiety/sleep medications. Clin Ther 1995;17:786-90.

10. Tanaka E, Hisawa S. Clinically significant pharmacokinetic drug interactions with psychoactive drugs: antidepressants and antipsychotics and the cytochrome P450 system. J Clin Pharm Ther 1999;24:7-16.

11. Bergendal L, Friberg A, Schaffrath A. Potential drug-drug interactions in 5,125 mostly elderly outpatients in Gothenburg, Sweden. Pharm World Sci 1995;17(5):152-7.

12. De Las Cuevas C, Sanz EJ. Polypharmacy in psychiatric practice in the Canary Islands. BMC Psychiatry 2004;4(1):18.-

13. Raschetti R, Morgutti M, Menniti Ippolito F, et al. Suspected adverse drug events requiring emergency department visits or hospital admissions. Eur J Clin Pharmacol 1999;54:959-63.

14. Bates DW, Spell N, Cullen DJ, et al. The costs of adverse drug events in hospitalized patients. JAMA 1997;277:307-11.

15. Frye MA, Ketter TA, Leverich GS, et al. The increasing use of polypharmacotherapy for refractory mood disorders: 22 years of study. J Clin Psychiatry 2000;1:9-15.

16. Werder SF, Preskorn SH. Managing polypharmacy: walking the fine line between help and harm. Current Psychiatry 2003;2(2):24-36.

17. Physicians’ Desk Reference (59th ed). Montvale, NJ: Thomson PDR; 2005.

18. Sandson NB. Exploring drug interaction in psychiatry. Psychiatric times 2004;May:42-8.

19. Todi SK, Hartmann RA. Pharmacologic principles. In: Civetta JM, Taylor RW, Kirby RR, (eds). Critical care (3rd ed). Philadelphia: Lippincott-Raven Publishers; 1997;485-8.

20. Shapiro LE, Shear NH. Drug interactions: Proteins, pumps, and P-450s. J Am Acad Dermatol 2002;47:467-84.

21. Keck PE, Jr, Dewan N, Nasrallah HA. Bipolar disorder: the clinician’s guide to pharmacotherapy for patients with co-occurring medical conditions. Current Psychiatry 2005;4(Feb)(suppl):1-51.

22. Sadock BJ, Sadock VA. Kaplan and Sadock’s pocket handbook of psychiatric drug treatment (3rd ed). Philadelphia: Lippincott Williams & Wilkins; 2001;144-5,178-83,256-7.

23. Shukla S, Godwin CD, Long LE, Miller MG. Lithium-carbamazepine neurotoxicity and risk factors. Am J Psychiatry 1984;141:1604-6.

24. Licht RW, Vestergaard P, Kessing LV, et al. Psychopharmacological treatment with lithium and antiepileptic drugs: suggested guidelines from the Danish Psychiatric Association and the Child and Adolescent Psychiatric Association in Denmark. Acta Psychiatr Scand Suppl 2003;(419):1-22.

25. Phelan KM, Mosholder AD, Lu S. Lithium interaction with the cyclooxygenase 2 inhibitors rofecoxib and celecoxib and other nonsteroidal anti-inflammatory drugs. J Clin Psychiatry 2003;64:1328-34.

26. Ragheb M, Ban TA, Buchanan D, Frolich JC. Interaction of indomethacin and ibuprofen with lithium in manic patients under steady-state lithium level. J Clin Psychiatry 1980;11:397-8.

Article PDF
Author and Disclosure Information

Mohamed Ibrahim Ramadan, MD
Chief resident

Steven F. Werder, DO
Assistant professor

Sheldon H. Preskorn, MD
Professor and chairman

Department of psychiatry and behavioral sciences, University of Kansas School of Medicine, Wichita

Issue
Current Psychiatry - 04(05)
Publications
Page Number
27-40
Sections
Author and Disclosure Information

Mohamed Ibrahim Ramadan, MD
Chief resident

Steven F. Werder, DO
Assistant professor

Sheldon H. Preskorn, MD
Professor and chairman

Department of psychiatry and behavioral sciences, University of Kansas School of Medicine, Wichita

Author and Disclosure Information

Mohamed Ibrahim Ramadan, MD
Chief resident

Steven F. Werder, DO
Assistant professor

Sheldon H. Preskorn, MD
Professor and chairman

Department of psychiatry and behavioral sciences, University of Kansas School of Medicine, Wichita

Article PDF
Article PDF

Drug-drug interactions (DDIs) can be viewed as physiologic combat wherein a “perpetrator” drug affects a “victim” drug’s pharmacokinetics or pharmacodynamics. Your challenge is to deter that interaction in patients taking two or more medications.

This article—first in a series—discusses polypharmacy risk factors that increase the likelihood of detrimental DDIs, then focuses on DDIs in patients taking mood stabilizers for bipolar disorder. We also offer practical tips to reduce DDI risk. Future articles will discuss DDI risks with antidepressants, antipsychotics, and anxiolytics.

To predict DDIs, you need to know psychotropics’ mechanism of action, metabolism, and effects on cytochrome P-450 (CYP) enzymes. Our discussion is not exhaustive because the data base is massive and new interactions continue to be discovered. Our aim is to equip you to anticipate and prevent DDIs when prescribing.

WHAT ARE ADVERSE DDIs?

An adverse event (AE) is any undesirable experience that occurs when a patient uses a medical product, whether or not the product caused the event. The FDA says an “undesirable experience” may be:

  • an unfavorable and unintended symptom or sign
  • an abnormal lab or radiographic finding
  • a disease that is temporarily associated with the medical product.
A temporal relationship is all that is required, although preexisting conditions and events clearly related to other causes are not usually considered adverse events.

An AE becomes “serious” (an SAE) when its duration, intensity, and/or frequency leads to death, a life-threatening condition, initial or prolonged hospitalization, disability, or congenital anomaly. Reporting is voluntary, but we strongly recommend that you report all SAEs to the FDA.

These definitions can help you confirm that a patient has experienced an SAE, but the task becomes more complicated when you try to attribute an SAE to a drug interaction. In the absence of an FDA definition, we assert that DDIs are responsible for SAEs when a perpetrator drug affects the pharmacokinetics or pharmacodynamics of a victim drug and exacerbates a known untoward event of the victim drug (Box 1).1-5 Which drug is the perpetrator and which is the victim is not always clear, and sometimes a medication—such as carbamazepine—can be both at once.

Box 1

Drug-drug interactions: Taking a toll

More than 100,000 possible detrimental DDIs have been documented in medical literature and pharmaceutical company data. This number is likely to grow with increased scrutiny, as

DDIs cause morbidity, mortality, and increased health care costs. More than 106,000 Americans die each year from properly prescribed, correctly taken medications. Polypharmacy is associated with extended hospital stays, and using >6 drugs is an independent predictor of death. DDIs contribute to the cause of death in acute overdoses and can be responsible for false-positive suicide diagnoses.

In clinical practice, DDI-associated toxicity may be mistaken for a new disease process, or a disease may be incorrectly perceived as progressing when a medication is rendered ineffective.

Source: References 1-5

RISKS OF POLYPHARMACY

Individuals with psychiatric illnesses are at particular risk for DDIs (Box 2). Patients seen by psychiatrists, for example, are six times more likely than patients seen by primary care physicians to be taking multiple medications.6

Polypharmacy increases the risk of adverse events, nonadherence, medication errors, and drug interactions.7 FDA’s MedWatch Web site lists more than 630 DDI warnings.8 The more medications a patient is taking, the greater the risk for detrimental DDIs and cumulative toxicity,9 which often lead to DDI-induced AEs.10

A study of DDIs in 5,125 mostly older outpatients11 found that:

  • 1,594 (31%) had at least one interacting drug combination (average 1.6)
  • subjects with one or more DDIs were taking an average 8.1 drugs, compared with 5.2 drugs in those without DDIs—a significant difference
  • 155 (3%) had interactions of “major clinical significance.”
‘Uncontrolled experiments.’ Drug combinations often are “uncontrolled experiments” with unknown potential for toxic effects.12 Studies have linked polypharmacy and DDIs as well as DDIs and AEs:

  • Although drug interactions are responsible for only 3.8% of emergency department visits, patients with DDIs are usually admitted to the hospital.13
  • Preventable drug interactions cause approximately one-third of all AEs in hospitalized patients and account for one-half of all AE costs.14
DDI risk is increasing over time as the number of medications used to treat psychiatric patients has grown. For example, 3.3% of patients discharged between 1974 and 1979 from the National Institute of Mental Health Biological Psychiatry Branch were taking 3 or more medications, compared with more than 40% of patients discharged between 1990 and 1995—a 12-fold increase.15

Box 2

Psychiatric patients: High risk for DDIs

Symptom-based prescribing. Patients with psychiatric illness are often prescribed >1 medication to manage symptoms and signs, rather than a single medication targeting a specific psychiatric disorder.

Multiple prescribers. Patients with anxiety and depressive disorders may see multiple providers, which increases the risk for polypharmacy, drug-drug interactions, and adverse events.

Medical comorbidity. Persons with psychiatric illness are at increased risk for concomitant medical illness, and persons with medical illness are at increased risk for psychiatric illness.

Psychiatric comorbidity. Persons with one psychiatric illness are at increased risk for other psychiatric illnesses.

Source: Adapted from reference 6.

 

 

HOW TO MINIMIZE DDI RISK

Use the acronym “LISTEN” (Table 1) to minimize DDI risk in patients taking combination therapies.16 The 6 steps in LISTEN can help you determine which drug or drugs you may discontinue before adding another.

We also recommend that you monitor therapeutic and toxic effects by checking serum drug levels, especially for drugs with a low therapeutic index. Lithium, for example, requires close mentoring of plasma concentration every 2 to 6 months and during dosage adjustments to avoid toxicity.17 Therapeutic drug monitoring has been shown to prevent adverse events from DDIs.16 For added safety, encourage patents to purchase all medications at one pharmacy and to enroll in that pharmacy’s DDI monitoring program.18

Keep in mind that systemic conditions may require a dosage change:

  • Increased volume of distribution, as in patients who gain weight or total water volume, requires higher doses to maintain a constant therapeutic effect.
  • Reduced clearance, as in patients with decreased renal or hepatic function, will likely require lower doses to prevent toxicity.19
Table 1

LISTEN: 6 tips to minimize DDI risk

LListeach drug’s name and dosage in the patient’s chart and in a note given to the patient.
IEach drug should have a clear indication and well-defined therapeutic goal; discontinue any drug not achieving its goal
SMake the regimen as simple as possible, with once- or twice-daily dosing.
TWhen possible, treat multiple symptoms with a single drug, rather than multiple symptoms with multiple drugs
EEducatepatients about polypharmacy, DDIs, and adverse events; assess all medications—including vitamins, minerals, herbs, dietary supplements, nonprescription products—and address potential DDIs
NAvoid prescribing medications with a narrow therapeutic window.

DDIsWITH MOOD STABILIZERS

Diagnoses of schizophrenia, anxiety disorders, and affective disorders are major risk factors for polypharmacy.20 DDIs are a particular concern in patients with bipolar disorder, given their complex treatment regimens.21

Interactions occur with the most commonly prescribed bipolar medications, including lithium and anticonvulsants (Table 2).17.21-25 Although atypical antipsychotics are also considered mood stabilizers in bipolar disorder, we will discuss their potential DDIs in a future article.

Table 2

Some drug-drug interactions with mood stabilizers

Mood stabilizerDrug interactions
Carbamazepine↑plasma clomipramine, phenytoin, primidone
↑risk of neurotoxic side effects and confusional states with lithium
Alters thyroid function with anticonvulsants
↓anticoagulant concentrations and↑bleeding risk
↓oral contraceptive reliability; can cause false-negative pregnancy tests
↑metabolism and may ↓efficacy of cancer chemotherapy (docetaxel, estrogens, paclitaxel, progesterone, cyclophosphamide)
↑aprepitant, granisetron metabolism and ↓efficacy
↑glipizide, tolbutamide metabolism
LithiumNSAIDs (ibuprofen, indomethacin, piroxicam) and COX-2 inhibitors ↑plasma lithium
ACE inhibitors ↑plasma lithium
Calcium channel blockers and carbamazepine ↑lithium neurotoxicity
SSRIs ↑diarrhea, confusion, tremor, dizziness, and agitation
Acetazolamide, urea, xanthine preparations, alkalinizing agents such as sodium bicarbonate ↓plasma lithium
Metronidazole ↑lithium toxicity
Encephalopathic syndrome possible with haloperidol
Lamotrigine↑concentration of carbamazepine’s epoxide metabolite
Carbamazepine, phenytoin, phenobarbital ↓plasma lamotrigine 40% to 50%
↑plasma sertraline
↓plasma valproic acid 25%; valproic acid doubles plasma lamotrigine and ↑rash risk
Topiramate↑valproic acid concentrations 11%; valproic acid ↓plasma topiramate 14%
↑plasma phenytoin up to 25%; phenytoin, carbamazepine ↓plasma topiramate by 40% to 48%
↓digoxin bioavailability
↓oral contraceptive efficacy
Valproic acid↑plasma phenobarbital, primidone
↓phenytoin clearance, volume distribution and ↑breakthrough seizure risk
↑serum concentration of antiepileptics, such as lamotrigine; absence status possible with clonazepam
↑= Increases ↓= Decreases
ACE = angiotensin-converting enzyme; COX = cyclooxygenase
NSAIDs = nonsteroidal anti-inflammatory drugs; SSRIs = selective serotonin reuptake inhibitors
Source: References 17, 21-25

LITHIUM: TOXICITY RISK

Lithium is excreted via the kidneys, so be cautious when using lithium in patients taking diuretics.17,22 Drugs that can lower serum lithium concentrations by increasing urinary lithium excretion include acetazolamide, urea, xanthine preparations, and alkalinizing agents such as sodium bicarbonate.17

Combining lithium with selective serotonin reuptake inhibitors can cause diarrhea, confusion, tremor, dizziness, and agitation.17 An encephalopathic syndrome has occurred in a few patients treated with lithium plus haloperidol.

Monitor lithium levels closely when bipolar patients start or stop nonsteroidal anti-inflammatory drugs (NSAIDs). Nonprescription ibuprofen can cause serious and even life-threatening serum lithium elevations by affecting lithium’s rate of tubular reabsorption.26 Indomethacin, piroxicam, and selective cyclooxygenase-2 (COX-2) inhibitors also increase plasma lithium concentrations.25

For patients taking lithium with heart drugs, angiotensin-converting enzyme (ACE) inhibitors may increase plasma lithium levels,17 and calcium channel blockers may increase the neurotoxicity risk.17,22 Using the anti-infective metronidazole with lithium may provoke lithium toxicity.

VALPROIC ACID: MONITOR CLEARANCE

Drugs that affect the expression of hepatic enzymes—especially glucuronosyltransferase—may increase clearance of valproic acid and its derivatives. Phenytoin, carbamazepine, or phenobarbital, for example, can double valproic acid clearance.

On the other hand, drugs that inhibit CYP-450 (such as antidepressants) have little effect on valproic acid concentration. Valproate can decrease plasma clearance of amitriptyline, so consider monitoring this tricyclic’s blood levels in patients also taking valproate.17

Because valproic acid can increase serum phenobarbital, monitor barbiturate concentrations when using these two drugs. A similar interaction occurs with primidone, which is metabolized into a barbiturate. Breakthrough seizures may occur with phenytoin, as valproic acid can reduce phenytoin clearance and apparent volume distribution by 25%.22

 

 

Using valproic acid with clonazepam may produce absence status in patients with a history of absence-type seizures.17 Valproic acid also displaces diazepam from its plasma albumin binding sites and inhibits its metabolism.

Concomitant use of valproic acid can increase serum concentrations of other antiepileptic drugs. For example, lamotrigine levels may double,24 and felbamate’s peak concentration may increase and require dosage reduction. Valproic acid may also interact with nonpsychiatric medications:

  • Subtherapeutic valproic acid levels have been reported when co-administered with the antibiotic meropenem.
  • In patients with HIV infection, valproic acid can decrease clearance of the antiretroviral zidovudine by 38%.
  • Patients receiving rifampin for tuberculosis may need a dosage adjustment, as oral rifampin’s clearance can increase 40% with concomitant valproic acid.

CARBAMAZEPINE: SELF-INDUCER

Metabolized by CYP 3A4, carbamazepine may induce its own metabolism as well as the CYP 3A4 isoenzyme. Therefore inhibitors and inducers of CYP 3A4 may affect carbamazepine plasma levels.

Carbamazepine can increase plasma levels of other psychotropics including clomipramine, phenytoin, and primidone.17,22 When used with lithium, it may increase the risk of neurotoxic side effects and confusion.23 It can alter thyroid function when used with other anticonvulsants.

For bipolar patients with diabetes, carbamazepine can cause hyperglycemia by inducing the metabolism of oral sulfonylureas such as glipizide and tolbutamide. In women, carbamazepine decreases the reliability of oral contraceptives17 and can cause false-negative pregnancy tests.24

For cancer patients, concurrent carbamazepine may induce metabolism of chemotherapy drugs such as docetaxel, estrogens, paclitaxel, progesterone, and cyclophosphamide, decreasing their efficacy.21 It can increase metabolism of aprepitant and granisetron—used to treat chemotherapy-related nausea—reducing plasma concentrations and possibly efficacy. Carbamazepine’s additive dopamine blockade can increase the risk of extrapyramidal symptoms when used with docetaxel or the antiemetic/antivertigo agents chlorpromazine, metoclopramide, or prochlorperazine.

Carbamazepine increases elimination of some cardiovascular drugs and may decrease the effect of antiarrhythmics such as lidocaine and quinidine; calcium channel blockers such as amlodipine, nifedipine, felodipine, nisoldipine, diltiazem, and verapamil; the beta blocker propranolol; and the vasodilator bosetan.21 Carbamazepine also reduces anticoagulant concentrations, and breakthrough bleeding has been reported.

(See "Out of the Pipeline−extended−release carbamazepine" for a listing of drugs that interact with this agent.)

OTHER ANTICONVULSANTS

Lamotrigine. Some concomitant CNS medications—such as carbamazepine, phenytoin or phenobarbital—reduce lamotrigine serum concentrations by as much as 50%.17 This substantial reduction may give the impression that the patient is not responding to therapeutic lamotrigine doses.

Patients taking lamotrigine with carbamazepine may be at greater risk for dizziness, diplopia, ataxia, and blurred vision because of increased serum concentration of carbamazepine’s epoxide metabolite. Valproic acid doubles lamotrigine serum concentration and increases the risk of rash, whereas lamotrigine decreases valproic acid concentration by 25%.17 Lamotrigine’s manufacturer offers special starting kits for patients taking carbamazepine or valproic acid.

Sertraline increases plasma lamotrigine concentration—but to a lesser extent than does valproic acid17 —and no dosage adjustment is needed.

Topiramate. Concomitant carbamazepine or phenytoin reduces topiramate concentration by 40% to 48%, whereas topiramate increases phenytoin concentration up to 25%. Similarly, valproic acid reduces topiramate’s concentration by 14%, while at the same time valproic acid concentration increases by 11%.17

Topiramate slightly decreases digoxin’s bioavailability and the efficacy of estrogenic oral contraceptives.17,22

Related resources

Drug brand names

  • Aprepitant • Emend
  • Bosentan • Tracleer
  • Carbamazepine • Tegretol, others
  • Chlorpromazine • Thorazine
  • Clomipramine • Anafranil, others
  • Clonazepam • Klonopin
  • Cyclophosphamide • Cytoxan, Neosar
  • Diazepam • Valium
  • Diltiazem • Cardizem, others
  • Docetaxel • Taxotere
  • Felbamate • Felbatol
  • Felodipine • Plendil
  • Granisetron • Kytril
  • Glipizide • Glucotrol
  • Haloperidol • Haldol
  • Indomethacin • Indocin
  • Lamotrigine • Lamictal
  • Meropenem • Merrem
  • Metoclopramide • Reglan
  • Metronidazole • Flagyl
  • Nifedipine • Adalat, Procardia
  • Nisoldipine • Sular
  • Paclitaxel • Taxol, others
  • Phenobarbital • Solfoton
  • Phenytoin • Dilantin
  • Piroxicam • Feldene
  • Primidone • Mysoline
  • Prochlorperazine • Compazine
  • Propranolol • Inderal
  • Rifampin • Rifadin
  • Sertraline • Zoloft
  • Tolbutamide • Orinase
  • Topiramate • Topamax
  • Valproic acid • Depakote
  • Verapamil • Calan, others
  • Zidovudine • Retrovir
Disclosure

Dr. Ramadan and Dr. Werder report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Preskorn has received grants or has been a consultant or speaker for Abbott Laboratories, AstraZeneca, Boehringer-Ingelheim, Bristol-Myers Squibb Co., Merck & Co., Eisai Inc., Eli Lilly and Co., GlaxoSmithKline, Janssen Pharmaceutica, Johnson & Johnson, Novartis Pharmaceuticals, Organon, Otsuka America Pharmaceutical Inc., Pfizer Inc., Solvay Pharmaceuticals, Sanofi-Aventis, and Wyeth.

Drug-drug interactions (DDIs) can be viewed as physiologic combat wherein a “perpetrator” drug affects a “victim” drug’s pharmacokinetics or pharmacodynamics. Your challenge is to deter that interaction in patients taking two or more medications.

This article—first in a series—discusses polypharmacy risk factors that increase the likelihood of detrimental DDIs, then focuses on DDIs in patients taking mood stabilizers for bipolar disorder. We also offer practical tips to reduce DDI risk. Future articles will discuss DDI risks with antidepressants, antipsychotics, and anxiolytics.

To predict DDIs, you need to know psychotropics’ mechanism of action, metabolism, and effects on cytochrome P-450 (CYP) enzymes. Our discussion is not exhaustive because the data base is massive and new interactions continue to be discovered. Our aim is to equip you to anticipate and prevent DDIs when prescribing.

WHAT ARE ADVERSE DDIs?

An adverse event (AE) is any undesirable experience that occurs when a patient uses a medical product, whether or not the product caused the event. The FDA says an “undesirable experience” may be:

  • an unfavorable and unintended symptom or sign
  • an abnormal lab or radiographic finding
  • a disease that is temporarily associated with the medical product.
A temporal relationship is all that is required, although preexisting conditions and events clearly related to other causes are not usually considered adverse events.

An AE becomes “serious” (an SAE) when its duration, intensity, and/or frequency leads to death, a life-threatening condition, initial or prolonged hospitalization, disability, or congenital anomaly. Reporting is voluntary, but we strongly recommend that you report all SAEs to the FDA.

These definitions can help you confirm that a patient has experienced an SAE, but the task becomes more complicated when you try to attribute an SAE to a drug interaction. In the absence of an FDA definition, we assert that DDIs are responsible for SAEs when a perpetrator drug affects the pharmacokinetics or pharmacodynamics of a victim drug and exacerbates a known untoward event of the victim drug (Box 1).1-5 Which drug is the perpetrator and which is the victim is not always clear, and sometimes a medication—such as carbamazepine—can be both at once.

Box 1

Drug-drug interactions: Taking a toll

More than 100,000 possible detrimental DDIs have been documented in medical literature and pharmaceutical company data. This number is likely to grow with increased scrutiny, as

DDIs cause morbidity, mortality, and increased health care costs. More than 106,000 Americans die each year from properly prescribed, correctly taken medications. Polypharmacy is associated with extended hospital stays, and using >6 drugs is an independent predictor of death. DDIs contribute to the cause of death in acute overdoses and can be responsible for false-positive suicide diagnoses.

In clinical practice, DDI-associated toxicity may be mistaken for a new disease process, or a disease may be incorrectly perceived as progressing when a medication is rendered ineffective.

Source: References 1-5

RISKS OF POLYPHARMACY

Individuals with psychiatric illnesses are at particular risk for DDIs (Box 2). Patients seen by psychiatrists, for example, are six times more likely than patients seen by primary care physicians to be taking multiple medications.6

Polypharmacy increases the risk of adverse events, nonadherence, medication errors, and drug interactions.7 FDA’s MedWatch Web site lists more than 630 DDI warnings.8 The more medications a patient is taking, the greater the risk for detrimental DDIs and cumulative toxicity,9 which often lead to DDI-induced AEs.10

A study of DDIs in 5,125 mostly older outpatients11 found that:

  • 1,594 (31%) had at least one interacting drug combination (average 1.6)
  • subjects with one or more DDIs were taking an average 8.1 drugs, compared with 5.2 drugs in those without DDIs—a significant difference
  • 155 (3%) had interactions of “major clinical significance.”
‘Uncontrolled experiments.’ Drug combinations often are “uncontrolled experiments” with unknown potential for toxic effects.12 Studies have linked polypharmacy and DDIs as well as DDIs and AEs:

  • Although drug interactions are responsible for only 3.8% of emergency department visits, patients with DDIs are usually admitted to the hospital.13
  • Preventable drug interactions cause approximately one-third of all AEs in hospitalized patients and account for one-half of all AE costs.14
DDI risk is increasing over time as the number of medications used to treat psychiatric patients has grown. For example, 3.3% of patients discharged between 1974 and 1979 from the National Institute of Mental Health Biological Psychiatry Branch were taking 3 or more medications, compared with more than 40% of patients discharged between 1990 and 1995—a 12-fold increase.15

Box 2

Psychiatric patients: High risk for DDIs

Symptom-based prescribing. Patients with psychiatric illness are often prescribed >1 medication to manage symptoms and signs, rather than a single medication targeting a specific psychiatric disorder.

Multiple prescribers. Patients with anxiety and depressive disorders may see multiple providers, which increases the risk for polypharmacy, drug-drug interactions, and adverse events.

Medical comorbidity. Persons with psychiatric illness are at increased risk for concomitant medical illness, and persons with medical illness are at increased risk for psychiatric illness.

Psychiatric comorbidity. Persons with one psychiatric illness are at increased risk for other psychiatric illnesses.

Source: Adapted from reference 6.

 

 

HOW TO MINIMIZE DDI RISK

Use the acronym “LISTEN” (Table 1) to minimize DDI risk in patients taking combination therapies.16 The 6 steps in LISTEN can help you determine which drug or drugs you may discontinue before adding another.

We also recommend that you monitor therapeutic and toxic effects by checking serum drug levels, especially for drugs with a low therapeutic index. Lithium, for example, requires close mentoring of plasma concentration every 2 to 6 months and during dosage adjustments to avoid toxicity.17 Therapeutic drug monitoring has been shown to prevent adverse events from DDIs.16 For added safety, encourage patents to purchase all medications at one pharmacy and to enroll in that pharmacy’s DDI monitoring program.18

Keep in mind that systemic conditions may require a dosage change:

  • Increased volume of distribution, as in patients who gain weight or total water volume, requires higher doses to maintain a constant therapeutic effect.
  • Reduced clearance, as in patients with decreased renal or hepatic function, will likely require lower doses to prevent toxicity.19
Table 1

LISTEN: 6 tips to minimize DDI risk

LListeach drug’s name and dosage in the patient’s chart and in a note given to the patient.
IEach drug should have a clear indication and well-defined therapeutic goal; discontinue any drug not achieving its goal
SMake the regimen as simple as possible, with once- or twice-daily dosing.
TWhen possible, treat multiple symptoms with a single drug, rather than multiple symptoms with multiple drugs
EEducatepatients about polypharmacy, DDIs, and adverse events; assess all medications—including vitamins, minerals, herbs, dietary supplements, nonprescription products—and address potential DDIs
NAvoid prescribing medications with a narrow therapeutic window.

DDIsWITH MOOD STABILIZERS

Diagnoses of schizophrenia, anxiety disorders, and affective disorders are major risk factors for polypharmacy.20 DDIs are a particular concern in patients with bipolar disorder, given their complex treatment regimens.21

Interactions occur with the most commonly prescribed bipolar medications, including lithium and anticonvulsants (Table 2).17.21-25 Although atypical antipsychotics are also considered mood stabilizers in bipolar disorder, we will discuss their potential DDIs in a future article.

Table 2

Some drug-drug interactions with mood stabilizers

Mood stabilizerDrug interactions
Carbamazepine↑plasma clomipramine, phenytoin, primidone
↑risk of neurotoxic side effects and confusional states with lithium
Alters thyroid function with anticonvulsants
↓anticoagulant concentrations and↑bleeding risk
↓oral contraceptive reliability; can cause false-negative pregnancy tests
↑metabolism and may ↓efficacy of cancer chemotherapy (docetaxel, estrogens, paclitaxel, progesterone, cyclophosphamide)
↑aprepitant, granisetron metabolism and ↓efficacy
↑glipizide, tolbutamide metabolism
LithiumNSAIDs (ibuprofen, indomethacin, piroxicam) and COX-2 inhibitors ↑plasma lithium
ACE inhibitors ↑plasma lithium
Calcium channel blockers and carbamazepine ↑lithium neurotoxicity
SSRIs ↑diarrhea, confusion, tremor, dizziness, and agitation
Acetazolamide, urea, xanthine preparations, alkalinizing agents such as sodium bicarbonate ↓plasma lithium
Metronidazole ↑lithium toxicity
Encephalopathic syndrome possible with haloperidol
Lamotrigine↑concentration of carbamazepine’s epoxide metabolite
Carbamazepine, phenytoin, phenobarbital ↓plasma lamotrigine 40% to 50%
↑plasma sertraline
↓plasma valproic acid 25%; valproic acid doubles plasma lamotrigine and ↑rash risk
Topiramate↑valproic acid concentrations 11%; valproic acid ↓plasma topiramate 14%
↑plasma phenytoin up to 25%; phenytoin, carbamazepine ↓plasma topiramate by 40% to 48%
↓digoxin bioavailability
↓oral contraceptive efficacy
Valproic acid↑plasma phenobarbital, primidone
↓phenytoin clearance, volume distribution and ↑breakthrough seizure risk
↑serum concentration of antiepileptics, such as lamotrigine; absence status possible with clonazepam
↑= Increases ↓= Decreases
ACE = angiotensin-converting enzyme; COX = cyclooxygenase
NSAIDs = nonsteroidal anti-inflammatory drugs; SSRIs = selective serotonin reuptake inhibitors
Source: References 17, 21-25

LITHIUM: TOXICITY RISK

Lithium is excreted via the kidneys, so be cautious when using lithium in patients taking diuretics.17,22 Drugs that can lower serum lithium concentrations by increasing urinary lithium excretion include acetazolamide, urea, xanthine preparations, and alkalinizing agents such as sodium bicarbonate.17

Combining lithium with selective serotonin reuptake inhibitors can cause diarrhea, confusion, tremor, dizziness, and agitation.17 An encephalopathic syndrome has occurred in a few patients treated with lithium plus haloperidol.

Monitor lithium levels closely when bipolar patients start or stop nonsteroidal anti-inflammatory drugs (NSAIDs). Nonprescription ibuprofen can cause serious and even life-threatening serum lithium elevations by affecting lithium’s rate of tubular reabsorption.26 Indomethacin, piroxicam, and selective cyclooxygenase-2 (COX-2) inhibitors also increase plasma lithium concentrations.25

For patients taking lithium with heart drugs, angiotensin-converting enzyme (ACE) inhibitors may increase plasma lithium levels,17 and calcium channel blockers may increase the neurotoxicity risk.17,22 Using the anti-infective metronidazole with lithium may provoke lithium toxicity.

VALPROIC ACID: MONITOR CLEARANCE

Drugs that affect the expression of hepatic enzymes—especially glucuronosyltransferase—may increase clearance of valproic acid and its derivatives. Phenytoin, carbamazepine, or phenobarbital, for example, can double valproic acid clearance.

On the other hand, drugs that inhibit CYP-450 (such as antidepressants) have little effect on valproic acid concentration. Valproate can decrease plasma clearance of amitriptyline, so consider monitoring this tricyclic’s blood levels in patients also taking valproate.17

Because valproic acid can increase serum phenobarbital, monitor barbiturate concentrations when using these two drugs. A similar interaction occurs with primidone, which is metabolized into a barbiturate. Breakthrough seizures may occur with phenytoin, as valproic acid can reduce phenytoin clearance and apparent volume distribution by 25%.22

 

 

Using valproic acid with clonazepam may produce absence status in patients with a history of absence-type seizures.17 Valproic acid also displaces diazepam from its plasma albumin binding sites and inhibits its metabolism.

Concomitant use of valproic acid can increase serum concentrations of other antiepileptic drugs. For example, lamotrigine levels may double,24 and felbamate’s peak concentration may increase and require dosage reduction. Valproic acid may also interact with nonpsychiatric medications:

  • Subtherapeutic valproic acid levels have been reported when co-administered with the antibiotic meropenem.
  • In patients with HIV infection, valproic acid can decrease clearance of the antiretroviral zidovudine by 38%.
  • Patients receiving rifampin for tuberculosis may need a dosage adjustment, as oral rifampin’s clearance can increase 40% with concomitant valproic acid.

CARBAMAZEPINE: SELF-INDUCER

Metabolized by CYP 3A4, carbamazepine may induce its own metabolism as well as the CYP 3A4 isoenzyme. Therefore inhibitors and inducers of CYP 3A4 may affect carbamazepine plasma levels.

Carbamazepine can increase plasma levels of other psychotropics including clomipramine, phenytoin, and primidone.17,22 When used with lithium, it may increase the risk of neurotoxic side effects and confusion.23 It can alter thyroid function when used with other anticonvulsants.

For bipolar patients with diabetes, carbamazepine can cause hyperglycemia by inducing the metabolism of oral sulfonylureas such as glipizide and tolbutamide. In women, carbamazepine decreases the reliability of oral contraceptives17 and can cause false-negative pregnancy tests.24

For cancer patients, concurrent carbamazepine may induce metabolism of chemotherapy drugs such as docetaxel, estrogens, paclitaxel, progesterone, and cyclophosphamide, decreasing their efficacy.21 It can increase metabolism of aprepitant and granisetron—used to treat chemotherapy-related nausea—reducing plasma concentrations and possibly efficacy. Carbamazepine’s additive dopamine blockade can increase the risk of extrapyramidal symptoms when used with docetaxel or the antiemetic/antivertigo agents chlorpromazine, metoclopramide, or prochlorperazine.

Carbamazepine increases elimination of some cardiovascular drugs and may decrease the effect of antiarrhythmics such as lidocaine and quinidine; calcium channel blockers such as amlodipine, nifedipine, felodipine, nisoldipine, diltiazem, and verapamil; the beta blocker propranolol; and the vasodilator bosetan.21 Carbamazepine also reduces anticoagulant concentrations, and breakthrough bleeding has been reported.

(See "Out of the Pipeline−extended−release carbamazepine" for a listing of drugs that interact with this agent.)

OTHER ANTICONVULSANTS

Lamotrigine. Some concomitant CNS medications—such as carbamazepine, phenytoin or phenobarbital—reduce lamotrigine serum concentrations by as much as 50%.17 This substantial reduction may give the impression that the patient is not responding to therapeutic lamotrigine doses.

Patients taking lamotrigine with carbamazepine may be at greater risk for dizziness, diplopia, ataxia, and blurred vision because of increased serum concentration of carbamazepine’s epoxide metabolite. Valproic acid doubles lamotrigine serum concentration and increases the risk of rash, whereas lamotrigine decreases valproic acid concentration by 25%.17 Lamotrigine’s manufacturer offers special starting kits for patients taking carbamazepine or valproic acid.

Sertraline increases plasma lamotrigine concentration—but to a lesser extent than does valproic acid17 —and no dosage adjustment is needed.

Topiramate. Concomitant carbamazepine or phenytoin reduces topiramate concentration by 40% to 48%, whereas topiramate increases phenytoin concentration up to 25%. Similarly, valproic acid reduces topiramate’s concentration by 14%, while at the same time valproic acid concentration increases by 11%.17

Topiramate slightly decreases digoxin’s bioavailability and the efficacy of estrogenic oral contraceptives.17,22

Related resources

Drug brand names

  • Aprepitant • Emend
  • Bosentan • Tracleer
  • Carbamazepine • Tegretol, others
  • Chlorpromazine • Thorazine
  • Clomipramine • Anafranil, others
  • Clonazepam • Klonopin
  • Cyclophosphamide • Cytoxan, Neosar
  • Diazepam • Valium
  • Diltiazem • Cardizem, others
  • Docetaxel • Taxotere
  • Felbamate • Felbatol
  • Felodipine • Plendil
  • Granisetron • Kytril
  • Glipizide • Glucotrol
  • Haloperidol • Haldol
  • Indomethacin • Indocin
  • Lamotrigine • Lamictal
  • Meropenem • Merrem
  • Metoclopramide • Reglan
  • Metronidazole • Flagyl
  • Nifedipine • Adalat, Procardia
  • Nisoldipine • Sular
  • Paclitaxel • Taxol, others
  • Phenobarbital • Solfoton
  • Phenytoin • Dilantin
  • Piroxicam • Feldene
  • Primidone • Mysoline
  • Prochlorperazine • Compazine
  • Propranolol • Inderal
  • Rifampin • Rifadin
  • Sertraline • Zoloft
  • Tolbutamide • Orinase
  • Topiramate • Topamax
  • Valproic acid • Depakote
  • Verapamil • Calan, others
  • Zidovudine • Retrovir
Disclosure

Dr. Ramadan and Dr. Werder report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Preskorn has received grants or has been a consultant or speaker for Abbott Laboratories, AstraZeneca, Boehringer-Ingelheim, Bristol-Myers Squibb Co., Merck & Co., Eisai Inc., Eli Lilly and Co., GlaxoSmithKline, Janssen Pharmaceutica, Johnson & Johnson, Novartis Pharmaceuticals, Organon, Otsuka America Pharmaceutical Inc., Pfizer Inc., Solvay Pharmaceuticals, Sanofi-Aventis, and Wyeth.

References

1. Langdorf MI, Fox JC, Marwah RS, et al. Physician versus computer knowledge of potential drug interactions in the emergency department. Acad Emerg Med 2000;7:1321-9.

2. Lazarou J, Pomeranz BH, Cory PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 1998;279(15):1200-5.

3. Incalzi RA, Gemma A, Capparella O, et al. Predicting mortality and length of stay of geriatric patients in an acute care general hospital. J Gerontol 1992;47(2):M35-9.

4. Preskorn SH. Fatal drug-drug interaction as a differential consideration in apparent suicide. J Psychiatr Pract 2002;8(4):233-8.

5. Peterson JF, Bates D. Preventable medication errors: identifying and eliminating serious drug interactions. J Am Pharm Assoc (Wash) 2001;41(2):159-60.

6. Nichol MB, Stimmel GL, Lange SC. Factors predicting the use of multiple psychotropic medications. J Clin Psychiatry 1995;2:60-6.

7. Ananth J, Parameswaran S, Gunatilake S. Antipsychotic polypharmacy. Curr Pharm Des 2004;10(18):2231-8.

8. Medwatch Web site. Food and Drug Administration. Search for Drug-drug interactions. Available at: http://www.fda.gov/med-watch/index.html. Accessed March 31, 2005.

9. Rascati K. Drug utilization review of concomitant use of specific serotonin reuptake inhibitors or clomipramine and antianxiety/sleep medications. Clin Ther 1995;17:786-90.

10. Tanaka E, Hisawa S. Clinically significant pharmacokinetic drug interactions with psychoactive drugs: antidepressants and antipsychotics and the cytochrome P450 system. J Clin Pharm Ther 1999;24:7-16.

11. Bergendal L, Friberg A, Schaffrath A. Potential drug-drug interactions in 5,125 mostly elderly outpatients in Gothenburg, Sweden. Pharm World Sci 1995;17(5):152-7.

12. De Las Cuevas C, Sanz EJ. Polypharmacy in psychiatric practice in the Canary Islands. BMC Psychiatry 2004;4(1):18.-

13. Raschetti R, Morgutti M, Menniti Ippolito F, et al. Suspected adverse drug events requiring emergency department visits or hospital admissions. Eur J Clin Pharmacol 1999;54:959-63.

14. Bates DW, Spell N, Cullen DJ, et al. The costs of adverse drug events in hospitalized patients. JAMA 1997;277:307-11.

15. Frye MA, Ketter TA, Leverich GS, et al. The increasing use of polypharmacotherapy for refractory mood disorders: 22 years of study. J Clin Psychiatry 2000;1:9-15.

16. Werder SF, Preskorn SH. Managing polypharmacy: walking the fine line between help and harm. Current Psychiatry 2003;2(2):24-36.

17. Physicians’ Desk Reference (59th ed). Montvale, NJ: Thomson PDR; 2005.

18. Sandson NB. Exploring drug interaction in psychiatry. Psychiatric times 2004;May:42-8.

19. Todi SK, Hartmann RA. Pharmacologic principles. In: Civetta JM, Taylor RW, Kirby RR, (eds). Critical care (3rd ed). Philadelphia: Lippincott-Raven Publishers; 1997;485-8.

20. Shapiro LE, Shear NH. Drug interactions: Proteins, pumps, and P-450s. J Am Acad Dermatol 2002;47:467-84.

21. Keck PE, Jr, Dewan N, Nasrallah HA. Bipolar disorder: the clinician’s guide to pharmacotherapy for patients with co-occurring medical conditions. Current Psychiatry 2005;4(Feb)(suppl):1-51.

22. Sadock BJ, Sadock VA. Kaplan and Sadock’s pocket handbook of psychiatric drug treatment (3rd ed). Philadelphia: Lippincott Williams & Wilkins; 2001;144-5,178-83,256-7.

23. Shukla S, Godwin CD, Long LE, Miller MG. Lithium-carbamazepine neurotoxicity and risk factors. Am J Psychiatry 1984;141:1604-6.

24. Licht RW, Vestergaard P, Kessing LV, et al. Psychopharmacological treatment with lithium and antiepileptic drugs: suggested guidelines from the Danish Psychiatric Association and the Child and Adolescent Psychiatric Association in Denmark. Acta Psychiatr Scand Suppl 2003;(419):1-22.

25. Phelan KM, Mosholder AD, Lu S. Lithium interaction with the cyclooxygenase 2 inhibitors rofecoxib and celecoxib and other nonsteroidal anti-inflammatory drugs. J Clin Psychiatry 2003;64:1328-34.

26. Ragheb M, Ban TA, Buchanan D, Frolich JC. Interaction of indomethacin and ibuprofen with lithium in manic patients under steady-state lithium level. J Clin Psychiatry 1980;11:397-8.

References

1. Langdorf MI, Fox JC, Marwah RS, et al. Physician versus computer knowledge of potential drug interactions in the emergency department. Acad Emerg Med 2000;7:1321-9.

2. Lazarou J, Pomeranz BH, Cory PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 1998;279(15):1200-5.

3. Incalzi RA, Gemma A, Capparella O, et al. Predicting mortality and length of stay of geriatric patients in an acute care general hospital. J Gerontol 1992;47(2):M35-9.

4. Preskorn SH. Fatal drug-drug interaction as a differential consideration in apparent suicide. J Psychiatr Pract 2002;8(4):233-8.

5. Peterson JF, Bates D. Preventable medication errors: identifying and eliminating serious drug interactions. J Am Pharm Assoc (Wash) 2001;41(2):159-60.

6. Nichol MB, Stimmel GL, Lange SC. Factors predicting the use of multiple psychotropic medications. J Clin Psychiatry 1995;2:60-6.

7. Ananth J, Parameswaran S, Gunatilake S. Antipsychotic polypharmacy. Curr Pharm Des 2004;10(18):2231-8.

8. Medwatch Web site. Food and Drug Administration. Search for Drug-drug interactions. Available at: http://www.fda.gov/med-watch/index.html. Accessed March 31, 2005.

9. Rascati K. Drug utilization review of concomitant use of specific serotonin reuptake inhibitors or clomipramine and antianxiety/sleep medications. Clin Ther 1995;17:786-90.

10. Tanaka E, Hisawa S. Clinically significant pharmacokinetic drug interactions with psychoactive drugs: antidepressants and antipsychotics and the cytochrome P450 system. J Clin Pharm Ther 1999;24:7-16.

11. Bergendal L, Friberg A, Schaffrath A. Potential drug-drug interactions in 5,125 mostly elderly outpatients in Gothenburg, Sweden. Pharm World Sci 1995;17(5):152-7.

12. De Las Cuevas C, Sanz EJ. Polypharmacy in psychiatric practice in the Canary Islands. BMC Psychiatry 2004;4(1):18.-

13. Raschetti R, Morgutti M, Menniti Ippolito F, et al. Suspected adverse drug events requiring emergency department visits or hospital admissions. Eur J Clin Pharmacol 1999;54:959-63.

14. Bates DW, Spell N, Cullen DJ, et al. The costs of adverse drug events in hospitalized patients. JAMA 1997;277:307-11.

15. Frye MA, Ketter TA, Leverich GS, et al. The increasing use of polypharmacotherapy for refractory mood disorders: 22 years of study. J Clin Psychiatry 2000;1:9-15.

16. Werder SF, Preskorn SH. Managing polypharmacy: walking the fine line between help and harm. Current Psychiatry 2003;2(2):24-36.

17. Physicians’ Desk Reference (59th ed). Montvale, NJ: Thomson PDR; 2005.

18. Sandson NB. Exploring drug interaction in psychiatry. Psychiatric times 2004;May:42-8.

19. Todi SK, Hartmann RA. Pharmacologic principles. In: Civetta JM, Taylor RW, Kirby RR, (eds). Critical care (3rd ed). Philadelphia: Lippincott-Raven Publishers; 1997;485-8.

20. Shapiro LE, Shear NH. Drug interactions: Proteins, pumps, and P-450s. J Am Acad Dermatol 2002;47:467-84.

21. Keck PE, Jr, Dewan N, Nasrallah HA. Bipolar disorder: the clinician’s guide to pharmacotherapy for patients with co-occurring medical conditions. Current Psychiatry 2005;4(Feb)(suppl):1-51.

22. Sadock BJ, Sadock VA. Kaplan and Sadock’s pocket handbook of psychiatric drug treatment (3rd ed). Philadelphia: Lippincott Williams & Wilkins; 2001;144-5,178-83,256-7.

23. Shukla S, Godwin CD, Long LE, Miller MG. Lithium-carbamazepine neurotoxicity and risk factors. Am J Psychiatry 1984;141:1604-6.

24. Licht RW, Vestergaard P, Kessing LV, et al. Psychopharmacological treatment with lithium and antiepileptic drugs: suggested guidelines from the Danish Psychiatric Association and the Child and Adolescent Psychiatric Association in Denmark. Acta Psychiatr Scand Suppl 2003;(419):1-22.

25. Phelan KM, Mosholder AD, Lu S. Lithium interaction with the cyclooxygenase 2 inhibitors rofecoxib and celecoxib and other nonsteroidal anti-inflammatory drugs. J Clin Psychiatry 2003;64:1328-34.

26. Ragheb M, Ban TA, Buchanan D, Frolich JC. Interaction of indomethacin and ibuprofen with lithium in manic patients under steady-state lithium level. J Clin Psychiatry 1980;11:397-8.

Issue
Current Psychiatry - 04(05)
Issue
Current Psychiatry - 04(05)
Page Number
27-40
Page Number
27-40
Publications
Publications
Article Type
Display Headline
Drug-drug interactions: Avoid serious adverse events with mood stabilizers
Display Headline
Drug-drug interactions: Avoid serious adverse events with mood stabilizers
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Something in the air

Article Type
Changed
Tue, 12/11/2018 - 15:31
Display Headline
Something in the air

HISTORY: WINTER WOES

Mrs. A, age 64, lives alone in an old farmhouse. For approximately 8 months, she had complained of depressed mood, decreased interest, difficulty sleeping, low energy, decreased concentration, and feelings of hopelessness. She met DSM-IV-TR criteria for major depressive disorder with underlying anxiety.

Mrs. A also reported having sinus headaches throughout the fall and winter. Blood chemistry, CBC with differential, thyroid profile including T4& TSH, urine drug screen, urine analysis, and ECG results were normal.

In April, Mrs. A was enrolled in an outpatient study of depression relapse prevention treatment. After taking the active study drug for 2 months, she reported continued low mood, low energy, difficulty concentrating, poor sleep and worsening headaches. Because her depression did not improve sufficiently, she was dropped from the study.

In July, Mrs. A saw a psychiatrist and was started on sertraline, 50 mg/d. By November, the dosage had been increased to 150 mg/d. At this time, she reported unsteadiness, dizziness, frequent falls, and intolerable headaches in addition to her depressive symptoms. She was referred to a neurologist to rule out a neurologic disorder.

Table 1

Symptoms that suggest major depression and/or chronic CO poisoning

SymptomMajor depressionChronic low-level CO poisoning
Depressed mood++
Diminished interest+-
Weight loss+-
Decreased appetite+-
Difficulty sleeping++
Diminished concentration++
Suicidal thoughts+-
Fatigue, weakness++
Headaches++
Palpitations++
Shortness of breath++
Nausea++
Abdominal pain++
Vomiting++
Diarrhea++
Confusion-+
Diminished cognitive function++
Sexual dysfunction+-
+ = suggests disorder
- = does not suggest disorder
CO = Carbon monoxide
Source: Diagnostic and Statistical Manual of Mental Disorders (4th ed, rev).
Copyright 2000. American Psychiatric Association; and Tierney LM, McPhee SJ, Papadakis MA (eds). Current Medical Diagnosis and Treatment. New York: McGraw Hill, 2003.

The authors’ observations

Chronic fatigue syndrome is characterized by severe unexplained fatigue that persists for >6 months. The new-onset fatigue is not abated with rest. Other symptoms include impaired memory or concentration, sore throat, tender lymph nodes, muscle pain, headaches, pain in several joints, and disturbed sleep.1

Mrs. A, however, never complained of sore throat or joint or muscle pain, and her laboratory findings were normal.

Seasonal affective disorder (SAD) is characterized by a temporal relationship between onset of depressive symptoms and a particular time of year (eg, symptoms emerge each winter) for at least 2 years. Full remission also occurs at a characteristic time (eg, each summer).2

Mrs. A’s headaches, frequent falls, dizziness, and difficulties with balance do not suggest SAD. Also, these symptoms have not persisted long enough for an SAD diagnosis.

Thyroid disorder. Hypothyroidism symptoms—particularly low mood, decreased energy, fatigue, psychomotor retardation, and lack of motivation—can mimic depression. Mrs. A’s T4 and TSH readings were normal, however.

Metabolic dysfunction. Symptoms secondary to decreased serum concentrations of sodium, potassium, magnesium, or calcium can mimic depression, but blood tests showed Mrs. A has normal electrolyte levels.

Brain tumor. Patients with a brain tumor can present with mood symptoms, psychosis, headaches, mania, cognitive impairments, seizure problems, and other symptoms depending on the tumor’s size and location.

FURTHER TREATMENT: SUDDEN RELIEF

By late November Mrs. A’s fatigue, once present only mornings, plagued her throughout the day. We considered changing antidepressants because of her complaints and sertraline’s lack of efficacy.

The following month, however, Mrs. A told us that her fatigue and headaches were gone. Mood, sleep, and concentration were also improved. Her Hamilton Rating Scale for Depression score had improved from 21 when she entered the study—indicating moderate severity—to 6, indicating remission. Her neurologic referral was cancelled.

Mrs. A then mentioned that her home’s water heater had been malfunctioning for several months. She said she could not afford to get it repaired during the summer but finally hired plumbers to fix it in late November.

After working all day in Mrs. A’s basement, two workers suffered acute headaches and nausea. The symptoms prompted the workers to search the basement for a carbon monoxide leak; they found a small leak in the water heater, which they replaced.

The next morning, Mrs. A said, her headache disappeared. Her other symptoms were gone within 4 days.

The authors’ observations

The sudden disappearance of Mrs. A’s symptoms after her water heater was replaced and emergence of severe physical symptoms in the two plumbers suggest carbon monoxide (CO) poisoning, a common and potentially lethal medical problem.

Low-level CO poisoning usually results from repeated exposure to incomplete combustion in a defective heating appliance, such as a water heater (Box 1).3,4 Symptoms usually surface in the winter, when heating appliance use peaks and windows are left closed, allowing indoor CO to accumulate in high concentrations.7

Box 1

Indoor heating systems: Leading cause of death by CO poisoning

Carbon monoxide (CO) poisoning is preventable yet causes more than 2,000 deaths each year in the United States.3,5 CO poisoning may result from intentional or accidental exposure to motor vehicle exhaust, malfunctioning home heating systems, and improperly vented combustion appliances.

Indoor heating systems account for about 75% of CO poisoning-related deaths.5 Fatal CO exposure has also been attributed to charcoal grills/burning charcoal, gas water heaters, camp stoves, lanterns, kitchen gas ranges/ovens, and other fuel-burning products.5

Although most states do not require residential use of CO detectors, clinicians should encourage patients to install at least one CO detector near their beds.5,6

 

 

Whereas severe, acute CO poisoning typically is detected immediately after exposure, symptoms of chronic low-level CO exposure are easily mistaken for a primary depressive (Table 1) or other neuropsychiatric disorder—or overlooked altogether. Some cases persist for months before CO exposure is diagnosed. Clinicians often give unnecessary—sometimes costly—medical treatment while ignoring the underlying poisoning.

Mechanism of action. CO binds with hemoglobin (with an affinity >200 times that of oxygen) to form carboxyhemoglobin (COHb), which causes cellular anoxia by blocking transport of oxygen to the tissues, including the brain.4,6,8

CO poisoning symptoms vary depending on COHb concentration (Table 2). COHb >5% in a symptomatic nonsmoker may indicate chronic low-level CO poisoning and require further evaluation.9 Levels >10% are common in heavy smokers (2 to 4 packs/day). It should be noted that Mrs. A does not smoke.

Presentation. Patients with chronic low-level CO poisoning often present with vague, nonspecific symptoms, such as weakness and fatigue, abdominal pain, nausea, vomiting, diarrhea, decreased concentration, diminished cognitive abilities, persistent headaches, and trouble sleeping.4,8,10,11 Patients age >65 especially may present with multiple cognitive and somatic complaints that suggest Parkinson’s disease, chronic fatigue syndrome, dementia, or—in Mrs. A’s case—depression.5,10,12

Table 2

Signs, symptoms of CO poisoning that emerge at different carboxyhemoglobin levels

Carboxyhemoglobin level (% HgB)Signs, symptoms
5-10 %Exacerbates angina in some patients with heart disease
10-20 %Mild headache, breathlessness on exertion
20-30 %Throbbing headache, irritability, mental status changes, fatigue
30-40 %Severe headache, weakness, nausea, dizziness, visual problems, confusion
40-50%Increased confusion, hallucinations, severe ataxia, rapid breathing
50-60 %Syncope or coma with convulsions, tachycardia with weak pulse
60-70 %Deep coma, incontinence
70-80%Profound coma, depressed respiration, absent reflexes
>80 %Rapid death from respiratory arrest
Source: Adapted from Gilman AG, Rall TW, Nies AS, Taylor P (eds). Goodman and Gilman’s the pharmacological basis of therapeutics (8th ed). New York: Pergamon Press, 1990.

Health effects of CO exposure range from subtle cardiovascular and neurobehavioral sequelae at low concentrations to loss of consciousness and death after acute exposure to higher concentrations.3,5

Hypoxia of the brain and other organs resulting from low-level CO poisoning can cause a range of physiologic effects, including mental status changes.10,11 Low-level CO exposure is particularly dangerous to pregnant women and to patients with a pre-existing ischemic illness.

Pregnancy. Chronic low-level CO exposure during pregnancy can harm the fetus, leading to low birth weight, short neonatal length, prematurity, perinatal death, and increased risk of developmental dysfunction.13

Ischemic illnesses. Because COHb cannot transport oxygen, the tissues that demand the most oxygen—such as the brain, heart, and skeletal muscles—are most affected. Because cardiac muscles extract approximately 75% of available oxygen from blood, patients with cardiac and pulmonary ischemic illnesses face a high risk for tissue injury with CO poisoning. At COHb levels >10%, patients with pre-existing cardiac disease experience increased severity and duration of angina; concentrations >15% place them at risk of myocardial infarction.6

Length of recovery from chronic CO exposure varies widely depending on severity of exposure and the patient’s general health.3,5 CO has a 4- to 6-hour half-life and is excreted via the lungs fairly rapidly, so recovery can be swift once CO exposure is stopped. Emergency room referral depends upon severity of symptoms and CO exposure duration and nature (accidental or intentional).

The authors’ observations

CO poisoning can lead to long-term mental status changes. In a 3-year follow-up of patients repeatedly exposed to low CO levels:

  • 43% developed neurologic sequelae including memory impairment
  • 33% experienced personality changes including irritability, verbal aggression, violence and impulsivity, moodiness, distractibility, and sexual promiscuity
  • 11% suffered gross neuropsychological effects, including psychosis, disorientation, and blindness.4

Primary care physicians and psychiatrists should monitor patients who have recovered from CO poisoning for symptoms of these disorders.

DETECTING CHRONIC CO EXPOSURE

Mrs. A’s case illustrates the seriousness and diagnostic complexity of chronic low-level CO exposure in older patients, especially during the fall and winter with increased home heating appliance use.7 CO exposure was not considered as a cause of Mrs. A’s symptoms until heating contractors found the water heater leak.

Watch for patients whose neuropsychiatric symptoms do not respond to treatment. Ask them about possible environmental, seasonal, or diurnal variations in symptoms. Also ask if the patient’s home heating system or water heater is ≥10 years old or has been malfunctioning (Box 2).

Checking COHb blood levels is the simplest way to confirm CO poisoning.6,14

Box 2

Seven questions to ask patients when you suspect chronic CO poisoning

  1. Is your home heating system or water heater 10 or more years old or malfunctioning?
  2. Do you use a gas range or stove for supplemental heat?
  3. Do symptoms improve or worsen in certain environments or at a certain time of day?
  4. Have fireplace flues and/or chimney vents been checked within the past year?
  5. Has another household member—including a pet—also been ill?
  6. Is a family member who remains at home persistently ill, whereas others who leave periodically improve?
  7. Do symptoms improve or worsen during certain months or seasons?
 

 

FOLLOW-UP: ANXIOUS MOMENTS

Mrs. A’s depressive symptoms, headaches, dizziness, and balance problems have not returned. Her underlying anxiety symptoms worsened, however, when the psychiatrist tried to taper sertraline. She was diagnosed with generalized anxiety disorder and continued on sertraline, 100 mg/d.

The psychiatrist sees her every 4 to 6 weeks, and she routinely sees her primary care physician. No long-term effects of CO poisoning have been found.

Related resources

  • U.S. Centers for Disease Control and Prevention. Enter “carbon monoxide poisoning” in search field. http://www.cdc.gov.
  • Kao LW, Nanagas KA. Carbon monoxide poisoning. Emerg Med Clin North America 2004;22:985-1018.

Drug brand names

  • Sertraline • Zoloft

Disclosure

Drs. Khan and D’Empaire report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Preskorn has been a speaker for, consultant to, or principal investigator for several antidepressant manufacturers, including Pfizer Inc.

References

1. Sadock BJ, Sadock VA. Kaplan and Sadock’s synopsis of psychiatry: behavioral sciences/clinical psychiatry (9th ed). Philadelphia, PA: Lippincott Williams & Wilkins, 2003:662.

2. Diagnostic and statistical manual of mental disorders (4th ed. rev). Washington, DC: American Psychiatric Association, 2000.

3. Mott JA, Wolfe MI, Alverson CJ, et al. National vehicle emissions policies and practices and declining US carbon monoxide-related mortality. JAMA 2002;288:988-95.

4. Thorpe M. Chronic carbon monoxide poisoning. Can J Psychiatry 1994;39:59-61.

5. Knobeloch L, Jackson R. Recognition of chronic carbon monoxide poisoning. WMJ 1999;98(6):26-9.

6. Turner M, Hamilton-Farrell MR, Clark RJ. Carbon monoxide poisoning: an update. J Accid Emerg Med 1999;16:92-6.

7. Unintentional carbon monoxide poisoning following winter storm—Washington January 1993. MMWR. 1993;42:109-11.

8. Wright J. Chronic and occult carbon monoxide poisoning: we don’t know what we’re missing. Emer Med J 2002;19:386-90.

9. Wald N, Idle M, Smith PG. Carboxyhaemoglobin levels in smokers of filter and plain cigarettes. Lancet 1977;1:110-12.

10. Raub JA, Benignus VA. Carbon monoxide and the nervous system. Neurosci Biobehav Rev 2002;26:925-40.

11. Ryan CM. Memory disturbances following chronic, low-level carbon monoxide exposure. Arch Clin Neuropsychol 1990;5:59-67.

12. Webb CJ, 2nd, Vaitkevicius PV. Dementia with a seasonal onset secondary to carbon monoxide poisoning. J Am Geriatr Soc 1997;45:1281-2.

13. Farrow JR, Davis GJ, Roy TM, et al. Fetal death due to nonlethal maternal carbon monoxide poisoning. J Forens Sci 1990;35:1448-52.

14. Vreman HJ, Mahoney JJ, Stevenson DK. Carbon monoxide and carboxyhemoglobin. Adv Pediatr 1995;42:303-34.

Author and Disclosure Information

Ahsan Y. Khan, MD
Assistant professor

Sheldon H. Preskorn, MD
Professor and chairman

Inna D’Empaire, MD
Third-year psychiatric resident

Department of psychiatry University of Kansas School of Medicine, Wichita

Issue
Current Psychiatry - 03(12)
Publications
Page Number
79-88
Sections
Author and Disclosure Information

Ahsan Y. Khan, MD
Assistant professor

Sheldon H. Preskorn, MD
Professor and chairman

Inna D’Empaire, MD
Third-year psychiatric resident

Department of psychiatry University of Kansas School of Medicine, Wichita

Author and Disclosure Information

Ahsan Y. Khan, MD
Assistant professor

Sheldon H. Preskorn, MD
Professor and chairman

Inna D’Empaire, MD
Third-year psychiatric resident

Department of psychiatry University of Kansas School of Medicine, Wichita

HISTORY: WINTER WOES

Mrs. A, age 64, lives alone in an old farmhouse. For approximately 8 months, she had complained of depressed mood, decreased interest, difficulty sleeping, low energy, decreased concentration, and feelings of hopelessness. She met DSM-IV-TR criteria for major depressive disorder with underlying anxiety.

Mrs. A also reported having sinus headaches throughout the fall and winter. Blood chemistry, CBC with differential, thyroid profile including T4& TSH, urine drug screen, urine analysis, and ECG results were normal.

In April, Mrs. A was enrolled in an outpatient study of depression relapse prevention treatment. After taking the active study drug for 2 months, she reported continued low mood, low energy, difficulty concentrating, poor sleep and worsening headaches. Because her depression did not improve sufficiently, she was dropped from the study.

In July, Mrs. A saw a psychiatrist and was started on sertraline, 50 mg/d. By November, the dosage had been increased to 150 mg/d. At this time, she reported unsteadiness, dizziness, frequent falls, and intolerable headaches in addition to her depressive symptoms. She was referred to a neurologist to rule out a neurologic disorder.

Table 1

Symptoms that suggest major depression and/or chronic CO poisoning

SymptomMajor depressionChronic low-level CO poisoning
Depressed mood++
Diminished interest+-
Weight loss+-
Decreased appetite+-
Difficulty sleeping++
Diminished concentration++
Suicidal thoughts+-
Fatigue, weakness++
Headaches++
Palpitations++
Shortness of breath++
Nausea++
Abdominal pain++
Vomiting++
Diarrhea++
Confusion-+
Diminished cognitive function++
Sexual dysfunction+-
+ = suggests disorder
- = does not suggest disorder
CO = Carbon monoxide
Source: Diagnostic and Statistical Manual of Mental Disorders (4th ed, rev).
Copyright 2000. American Psychiatric Association; and Tierney LM, McPhee SJ, Papadakis MA (eds). Current Medical Diagnosis and Treatment. New York: McGraw Hill, 2003.

The authors’ observations

Chronic fatigue syndrome is characterized by severe unexplained fatigue that persists for >6 months. The new-onset fatigue is not abated with rest. Other symptoms include impaired memory or concentration, sore throat, tender lymph nodes, muscle pain, headaches, pain in several joints, and disturbed sleep.1

Mrs. A, however, never complained of sore throat or joint or muscle pain, and her laboratory findings were normal.

Seasonal affective disorder (SAD) is characterized by a temporal relationship between onset of depressive symptoms and a particular time of year (eg, symptoms emerge each winter) for at least 2 years. Full remission also occurs at a characteristic time (eg, each summer).2

Mrs. A’s headaches, frequent falls, dizziness, and difficulties with balance do not suggest SAD. Also, these symptoms have not persisted long enough for an SAD diagnosis.

Thyroid disorder. Hypothyroidism symptoms—particularly low mood, decreased energy, fatigue, psychomotor retardation, and lack of motivation—can mimic depression. Mrs. A’s T4 and TSH readings were normal, however.

Metabolic dysfunction. Symptoms secondary to decreased serum concentrations of sodium, potassium, magnesium, or calcium can mimic depression, but blood tests showed Mrs. A has normal electrolyte levels.

Brain tumor. Patients with a brain tumor can present with mood symptoms, psychosis, headaches, mania, cognitive impairments, seizure problems, and other symptoms depending on the tumor’s size and location.

FURTHER TREATMENT: SUDDEN RELIEF

By late November Mrs. A’s fatigue, once present only mornings, plagued her throughout the day. We considered changing antidepressants because of her complaints and sertraline’s lack of efficacy.

The following month, however, Mrs. A told us that her fatigue and headaches were gone. Mood, sleep, and concentration were also improved. Her Hamilton Rating Scale for Depression score had improved from 21 when she entered the study—indicating moderate severity—to 6, indicating remission. Her neurologic referral was cancelled.

Mrs. A then mentioned that her home’s water heater had been malfunctioning for several months. She said she could not afford to get it repaired during the summer but finally hired plumbers to fix it in late November.

After working all day in Mrs. A’s basement, two workers suffered acute headaches and nausea. The symptoms prompted the workers to search the basement for a carbon monoxide leak; they found a small leak in the water heater, which they replaced.

The next morning, Mrs. A said, her headache disappeared. Her other symptoms were gone within 4 days.

The authors’ observations

The sudden disappearance of Mrs. A’s symptoms after her water heater was replaced and emergence of severe physical symptoms in the two plumbers suggest carbon monoxide (CO) poisoning, a common and potentially lethal medical problem.

Low-level CO poisoning usually results from repeated exposure to incomplete combustion in a defective heating appliance, such as a water heater (Box 1).3,4 Symptoms usually surface in the winter, when heating appliance use peaks and windows are left closed, allowing indoor CO to accumulate in high concentrations.7

Box 1

Indoor heating systems: Leading cause of death by CO poisoning

Carbon monoxide (CO) poisoning is preventable yet causes more than 2,000 deaths each year in the United States.3,5 CO poisoning may result from intentional or accidental exposure to motor vehicle exhaust, malfunctioning home heating systems, and improperly vented combustion appliances.

Indoor heating systems account for about 75% of CO poisoning-related deaths.5 Fatal CO exposure has also been attributed to charcoal grills/burning charcoal, gas water heaters, camp stoves, lanterns, kitchen gas ranges/ovens, and other fuel-burning products.5

Although most states do not require residential use of CO detectors, clinicians should encourage patients to install at least one CO detector near their beds.5,6

 

 

Whereas severe, acute CO poisoning typically is detected immediately after exposure, symptoms of chronic low-level CO exposure are easily mistaken for a primary depressive (Table 1) or other neuropsychiatric disorder—or overlooked altogether. Some cases persist for months before CO exposure is diagnosed. Clinicians often give unnecessary—sometimes costly—medical treatment while ignoring the underlying poisoning.

Mechanism of action. CO binds with hemoglobin (with an affinity >200 times that of oxygen) to form carboxyhemoglobin (COHb), which causes cellular anoxia by blocking transport of oxygen to the tissues, including the brain.4,6,8

CO poisoning symptoms vary depending on COHb concentration (Table 2). COHb >5% in a symptomatic nonsmoker may indicate chronic low-level CO poisoning and require further evaluation.9 Levels >10% are common in heavy smokers (2 to 4 packs/day). It should be noted that Mrs. A does not smoke.

Presentation. Patients with chronic low-level CO poisoning often present with vague, nonspecific symptoms, such as weakness and fatigue, abdominal pain, nausea, vomiting, diarrhea, decreased concentration, diminished cognitive abilities, persistent headaches, and trouble sleeping.4,8,10,11 Patients age >65 especially may present with multiple cognitive and somatic complaints that suggest Parkinson’s disease, chronic fatigue syndrome, dementia, or—in Mrs. A’s case—depression.5,10,12

Table 2

Signs, symptoms of CO poisoning that emerge at different carboxyhemoglobin levels

Carboxyhemoglobin level (% HgB)Signs, symptoms
5-10 %Exacerbates angina in some patients with heart disease
10-20 %Mild headache, breathlessness on exertion
20-30 %Throbbing headache, irritability, mental status changes, fatigue
30-40 %Severe headache, weakness, nausea, dizziness, visual problems, confusion
40-50%Increased confusion, hallucinations, severe ataxia, rapid breathing
50-60 %Syncope or coma with convulsions, tachycardia with weak pulse
60-70 %Deep coma, incontinence
70-80%Profound coma, depressed respiration, absent reflexes
>80 %Rapid death from respiratory arrest
Source: Adapted from Gilman AG, Rall TW, Nies AS, Taylor P (eds). Goodman and Gilman’s the pharmacological basis of therapeutics (8th ed). New York: Pergamon Press, 1990.

Health effects of CO exposure range from subtle cardiovascular and neurobehavioral sequelae at low concentrations to loss of consciousness and death after acute exposure to higher concentrations.3,5

Hypoxia of the brain and other organs resulting from low-level CO poisoning can cause a range of physiologic effects, including mental status changes.10,11 Low-level CO exposure is particularly dangerous to pregnant women and to patients with a pre-existing ischemic illness.

Pregnancy. Chronic low-level CO exposure during pregnancy can harm the fetus, leading to low birth weight, short neonatal length, prematurity, perinatal death, and increased risk of developmental dysfunction.13

Ischemic illnesses. Because COHb cannot transport oxygen, the tissues that demand the most oxygen—such as the brain, heart, and skeletal muscles—are most affected. Because cardiac muscles extract approximately 75% of available oxygen from blood, patients with cardiac and pulmonary ischemic illnesses face a high risk for tissue injury with CO poisoning. At COHb levels >10%, patients with pre-existing cardiac disease experience increased severity and duration of angina; concentrations >15% place them at risk of myocardial infarction.6

Length of recovery from chronic CO exposure varies widely depending on severity of exposure and the patient’s general health.3,5 CO has a 4- to 6-hour half-life and is excreted via the lungs fairly rapidly, so recovery can be swift once CO exposure is stopped. Emergency room referral depends upon severity of symptoms and CO exposure duration and nature (accidental or intentional).

The authors’ observations

CO poisoning can lead to long-term mental status changes. In a 3-year follow-up of patients repeatedly exposed to low CO levels:

  • 43% developed neurologic sequelae including memory impairment
  • 33% experienced personality changes including irritability, verbal aggression, violence and impulsivity, moodiness, distractibility, and sexual promiscuity
  • 11% suffered gross neuropsychological effects, including psychosis, disorientation, and blindness.4

Primary care physicians and psychiatrists should monitor patients who have recovered from CO poisoning for symptoms of these disorders.

DETECTING CHRONIC CO EXPOSURE

Mrs. A’s case illustrates the seriousness and diagnostic complexity of chronic low-level CO exposure in older patients, especially during the fall and winter with increased home heating appliance use.7 CO exposure was not considered as a cause of Mrs. A’s symptoms until heating contractors found the water heater leak.

Watch for patients whose neuropsychiatric symptoms do not respond to treatment. Ask them about possible environmental, seasonal, or diurnal variations in symptoms. Also ask if the patient’s home heating system or water heater is ≥10 years old or has been malfunctioning (Box 2).

Checking COHb blood levels is the simplest way to confirm CO poisoning.6,14

Box 2

Seven questions to ask patients when you suspect chronic CO poisoning

  1. Is your home heating system or water heater 10 or more years old or malfunctioning?
  2. Do you use a gas range or stove for supplemental heat?
  3. Do symptoms improve or worsen in certain environments or at a certain time of day?
  4. Have fireplace flues and/or chimney vents been checked within the past year?
  5. Has another household member—including a pet—also been ill?
  6. Is a family member who remains at home persistently ill, whereas others who leave periodically improve?
  7. Do symptoms improve or worsen during certain months or seasons?
 

 

FOLLOW-UP: ANXIOUS MOMENTS

Mrs. A’s depressive symptoms, headaches, dizziness, and balance problems have not returned. Her underlying anxiety symptoms worsened, however, when the psychiatrist tried to taper sertraline. She was diagnosed with generalized anxiety disorder and continued on sertraline, 100 mg/d.

The psychiatrist sees her every 4 to 6 weeks, and she routinely sees her primary care physician. No long-term effects of CO poisoning have been found.

Related resources

  • U.S. Centers for Disease Control and Prevention. Enter “carbon monoxide poisoning” in search field. http://www.cdc.gov.
  • Kao LW, Nanagas KA. Carbon monoxide poisoning. Emerg Med Clin North America 2004;22:985-1018.

Drug brand names

  • Sertraline • Zoloft

Disclosure

Drs. Khan and D’Empaire report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Preskorn has been a speaker for, consultant to, or principal investigator for several antidepressant manufacturers, including Pfizer Inc.

HISTORY: WINTER WOES

Mrs. A, age 64, lives alone in an old farmhouse. For approximately 8 months, she had complained of depressed mood, decreased interest, difficulty sleeping, low energy, decreased concentration, and feelings of hopelessness. She met DSM-IV-TR criteria for major depressive disorder with underlying anxiety.

Mrs. A also reported having sinus headaches throughout the fall and winter. Blood chemistry, CBC with differential, thyroid profile including T4& TSH, urine drug screen, urine analysis, and ECG results were normal.

In April, Mrs. A was enrolled in an outpatient study of depression relapse prevention treatment. After taking the active study drug for 2 months, she reported continued low mood, low energy, difficulty concentrating, poor sleep and worsening headaches. Because her depression did not improve sufficiently, she was dropped from the study.

In July, Mrs. A saw a psychiatrist and was started on sertraline, 50 mg/d. By November, the dosage had been increased to 150 mg/d. At this time, she reported unsteadiness, dizziness, frequent falls, and intolerable headaches in addition to her depressive symptoms. She was referred to a neurologist to rule out a neurologic disorder.

Table 1

Symptoms that suggest major depression and/or chronic CO poisoning

SymptomMajor depressionChronic low-level CO poisoning
Depressed mood++
Diminished interest+-
Weight loss+-
Decreased appetite+-
Difficulty sleeping++
Diminished concentration++
Suicidal thoughts+-
Fatigue, weakness++
Headaches++
Palpitations++
Shortness of breath++
Nausea++
Abdominal pain++
Vomiting++
Diarrhea++
Confusion-+
Diminished cognitive function++
Sexual dysfunction+-
+ = suggests disorder
- = does not suggest disorder
CO = Carbon monoxide
Source: Diagnostic and Statistical Manual of Mental Disorders (4th ed, rev).
Copyright 2000. American Psychiatric Association; and Tierney LM, McPhee SJ, Papadakis MA (eds). Current Medical Diagnosis and Treatment. New York: McGraw Hill, 2003.

The authors’ observations

Chronic fatigue syndrome is characterized by severe unexplained fatigue that persists for >6 months. The new-onset fatigue is not abated with rest. Other symptoms include impaired memory or concentration, sore throat, tender lymph nodes, muscle pain, headaches, pain in several joints, and disturbed sleep.1

Mrs. A, however, never complained of sore throat or joint or muscle pain, and her laboratory findings were normal.

Seasonal affective disorder (SAD) is characterized by a temporal relationship between onset of depressive symptoms and a particular time of year (eg, symptoms emerge each winter) for at least 2 years. Full remission also occurs at a characteristic time (eg, each summer).2

Mrs. A’s headaches, frequent falls, dizziness, and difficulties with balance do not suggest SAD. Also, these symptoms have not persisted long enough for an SAD diagnosis.

Thyroid disorder. Hypothyroidism symptoms—particularly low mood, decreased energy, fatigue, psychomotor retardation, and lack of motivation—can mimic depression. Mrs. A’s T4 and TSH readings were normal, however.

Metabolic dysfunction. Symptoms secondary to decreased serum concentrations of sodium, potassium, magnesium, or calcium can mimic depression, but blood tests showed Mrs. A has normal electrolyte levels.

Brain tumor. Patients with a brain tumor can present with mood symptoms, psychosis, headaches, mania, cognitive impairments, seizure problems, and other symptoms depending on the tumor’s size and location.

FURTHER TREATMENT: SUDDEN RELIEF

By late November Mrs. A’s fatigue, once present only mornings, plagued her throughout the day. We considered changing antidepressants because of her complaints and sertraline’s lack of efficacy.

The following month, however, Mrs. A told us that her fatigue and headaches were gone. Mood, sleep, and concentration were also improved. Her Hamilton Rating Scale for Depression score had improved from 21 when she entered the study—indicating moderate severity—to 6, indicating remission. Her neurologic referral was cancelled.

Mrs. A then mentioned that her home’s water heater had been malfunctioning for several months. She said she could not afford to get it repaired during the summer but finally hired plumbers to fix it in late November.

After working all day in Mrs. A’s basement, two workers suffered acute headaches and nausea. The symptoms prompted the workers to search the basement for a carbon monoxide leak; they found a small leak in the water heater, which they replaced.

The next morning, Mrs. A said, her headache disappeared. Her other symptoms were gone within 4 days.

The authors’ observations

The sudden disappearance of Mrs. A’s symptoms after her water heater was replaced and emergence of severe physical symptoms in the two plumbers suggest carbon monoxide (CO) poisoning, a common and potentially lethal medical problem.

Low-level CO poisoning usually results from repeated exposure to incomplete combustion in a defective heating appliance, such as a water heater (Box 1).3,4 Symptoms usually surface in the winter, when heating appliance use peaks and windows are left closed, allowing indoor CO to accumulate in high concentrations.7

Box 1

Indoor heating systems: Leading cause of death by CO poisoning

Carbon monoxide (CO) poisoning is preventable yet causes more than 2,000 deaths each year in the United States.3,5 CO poisoning may result from intentional or accidental exposure to motor vehicle exhaust, malfunctioning home heating systems, and improperly vented combustion appliances.

Indoor heating systems account for about 75% of CO poisoning-related deaths.5 Fatal CO exposure has also been attributed to charcoal grills/burning charcoal, gas water heaters, camp stoves, lanterns, kitchen gas ranges/ovens, and other fuel-burning products.5

Although most states do not require residential use of CO detectors, clinicians should encourage patients to install at least one CO detector near their beds.5,6

 

 

Whereas severe, acute CO poisoning typically is detected immediately after exposure, symptoms of chronic low-level CO exposure are easily mistaken for a primary depressive (Table 1) or other neuropsychiatric disorder—or overlooked altogether. Some cases persist for months before CO exposure is diagnosed. Clinicians often give unnecessary—sometimes costly—medical treatment while ignoring the underlying poisoning.

Mechanism of action. CO binds with hemoglobin (with an affinity >200 times that of oxygen) to form carboxyhemoglobin (COHb), which causes cellular anoxia by blocking transport of oxygen to the tissues, including the brain.4,6,8

CO poisoning symptoms vary depending on COHb concentration (Table 2). COHb >5% in a symptomatic nonsmoker may indicate chronic low-level CO poisoning and require further evaluation.9 Levels >10% are common in heavy smokers (2 to 4 packs/day). It should be noted that Mrs. A does not smoke.

Presentation. Patients with chronic low-level CO poisoning often present with vague, nonspecific symptoms, such as weakness and fatigue, abdominal pain, nausea, vomiting, diarrhea, decreased concentration, diminished cognitive abilities, persistent headaches, and trouble sleeping.4,8,10,11 Patients age >65 especially may present with multiple cognitive and somatic complaints that suggest Parkinson’s disease, chronic fatigue syndrome, dementia, or—in Mrs. A’s case—depression.5,10,12

Table 2

Signs, symptoms of CO poisoning that emerge at different carboxyhemoglobin levels

Carboxyhemoglobin level (% HgB)Signs, symptoms
5-10 %Exacerbates angina in some patients with heart disease
10-20 %Mild headache, breathlessness on exertion
20-30 %Throbbing headache, irritability, mental status changes, fatigue
30-40 %Severe headache, weakness, nausea, dizziness, visual problems, confusion
40-50%Increased confusion, hallucinations, severe ataxia, rapid breathing
50-60 %Syncope or coma with convulsions, tachycardia with weak pulse
60-70 %Deep coma, incontinence
70-80%Profound coma, depressed respiration, absent reflexes
>80 %Rapid death from respiratory arrest
Source: Adapted from Gilman AG, Rall TW, Nies AS, Taylor P (eds). Goodman and Gilman’s the pharmacological basis of therapeutics (8th ed). New York: Pergamon Press, 1990.

Health effects of CO exposure range from subtle cardiovascular and neurobehavioral sequelae at low concentrations to loss of consciousness and death after acute exposure to higher concentrations.3,5

Hypoxia of the brain and other organs resulting from low-level CO poisoning can cause a range of physiologic effects, including mental status changes.10,11 Low-level CO exposure is particularly dangerous to pregnant women and to patients with a pre-existing ischemic illness.

Pregnancy. Chronic low-level CO exposure during pregnancy can harm the fetus, leading to low birth weight, short neonatal length, prematurity, perinatal death, and increased risk of developmental dysfunction.13

Ischemic illnesses. Because COHb cannot transport oxygen, the tissues that demand the most oxygen—such as the brain, heart, and skeletal muscles—are most affected. Because cardiac muscles extract approximately 75% of available oxygen from blood, patients with cardiac and pulmonary ischemic illnesses face a high risk for tissue injury with CO poisoning. At COHb levels >10%, patients with pre-existing cardiac disease experience increased severity and duration of angina; concentrations >15% place them at risk of myocardial infarction.6

Length of recovery from chronic CO exposure varies widely depending on severity of exposure and the patient’s general health.3,5 CO has a 4- to 6-hour half-life and is excreted via the lungs fairly rapidly, so recovery can be swift once CO exposure is stopped. Emergency room referral depends upon severity of symptoms and CO exposure duration and nature (accidental or intentional).

The authors’ observations

CO poisoning can lead to long-term mental status changes. In a 3-year follow-up of patients repeatedly exposed to low CO levels:

  • 43% developed neurologic sequelae including memory impairment
  • 33% experienced personality changes including irritability, verbal aggression, violence and impulsivity, moodiness, distractibility, and sexual promiscuity
  • 11% suffered gross neuropsychological effects, including psychosis, disorientation, and blindness.4

Primary care physicians and psychiatrists should monitor patients who have recovered from CO poisoning for symptoms of these disorders.

DETECTING CHRONIC CO EXPOSURE

Mrs. A’s case illustrates the seriousness and diagnostic complexity of chronic low-level CO exposure in older patients, especially during the fall and winter with increased home heating appliance use.7 CO exposure was not considered as a cause of Mrs. A’s symptoms until heating contractors found the water heater leak.

Watch for patients whose neuropsychiatric symptoms do not respond to treatment. Ask them about possible environmental, seasonal, or diurnal variations in symptoms. Also ask if the patient’s home heating system or water heater is ≥10 years old or has been malfunctioning (Box 2).

Checking COHb blood levels is the simplest way to confirm CO poisoning.6,14

Box 2

Seven questions to ask patients when you suspect chronic CO poisoning

  1. Is your home heating system or water heater 10 or more years old or malfunctioning?
  2. Do you use a gas range or stove for supplemental heat?
  3. Do symptoms improve or worsen in certain environments or at a certain time of day?
  4. Have fireplace flues and/or chimney vents been checked within the past year?
  5. Has another household member—including a pet—also been ill?
  6. Is a family member who remains at home persistently ill, whereas others who leave periodically improve?
  7. Do symptoms improve or worsen during certain months or seasons?
 

 

FOLLOW-UP: ANXIOUS MOMENTS

Mrs. A’s depressive symptoms, headaches, dizziness, and balance problems have not returned. Her underlying anxiety symptoms worsened, however, when the psychiatrist tried to taper sertraline. She was diagnosed with generalized anxiety disorder and continued on sertraline, 100 mg/d.

The psychiatrist sees her every 4 to 6 weeks, and she routinely sees her primary care physician. No long-term effects of CO poisoning have been found.

Related resources

  • U.S. Centers for Disease Control and Prevention. Enter “carbon monoxide poisoning” in search field. http://www.cdc.gov.
  • Kao LW, Nanagas KA. Carbon monoxide poisoning. Emerg Med Clin North America 2004;22:985-1018.

Drug brand names

  • Sertraline • Zoloft

Disclosure

Drs. Khan and D’Empaire report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Preskorn has been a speaker for, consultant to, or principal investigator for several antidepressant manufacturers, including Pfizer Inc.

References

1. Sadock BJ, Sadock VA. Kaplan and Sadock’s synopsis of psychiatry: behavioral sciences/clinical psychiatry (9th ed). Philadelphia, PA: Lippincott Williams & Wilkins, 2003:662.

2. Diagnostic and statistical manual of mental disorders (4th ed. rev). Washington, DC: American Psychiatric Association, 2000.

3. Mott JA, Wolfe MI, Alverson CJ, et al. National vehicle emissions policies and practices and declining US carbon monoxide-related mortality. JAMA 2002;288:988-95.

4. Thorpe M. Chronic carbon monoxide poisoning. Can J Psychiatry 1994;39:59-61.

5. Knobeloch L, Jackson R. Recognition of chronic carbon monoxide poisoning. WMJ 1999;98(6):26-9.

6. Turner M, Hamilton-Farrell MR, Clark RJ. Carbon monoxide poisoning: an update. J Accid Emerg Med 1999;16:92-6.

7. Unintentional carbon monoxide poisoning following winter storm—Washington January 1993. MMWR. 1993;42:109-11.

8. Wright J. Chronic and occult carbon monoxide poisoning: we don’t know what we’re missing. Emer Med J 2002;19:386-90.

9. Wald N, Idle M, Smith PG. Carboxyhaemoglobin levels in smokers of filter and plain cigarettes. Lancet 1977;1:110-12.

10. Raub JA, Benignus VA. Carbon monoxide and the nervous system. Neurosci Biobehav Rev 2002;26:925-40.

11. Ryan CM. Memory disturbances following chronic, low-level carbon monoxide exposure. Arch Clin Neuropsychol 1990;5:59-67.

12. Webb CJ, 2nd, Vaitkevicius PV. Dementia with a seasonal onset secondary to carbon monoxide poisoning. J Am Geriatr Soc 1997;45:1281-2.

13. Farrow JR, Davis GJ, Roy TM, et al. Fetal death due to nonlethal maternal carbon monoxide poisoning. J Forens Sci 1990;35:1448-52.

14. Vreman HJ, Mahoney JJ, Stevenson DK. Carbon monoxide and carboxyhemoglobin. Adv Pediatr 1995;42:303-34.

References

1. Sadock BJ, Sadock VA. Kaplan and Sadock’s synopsis of psychiatry: behavioral sciences/clinical psychiatry (9th ed). Philadelphia, PA: Lippincott Williams & Wilkins, 2003:662.

2. Diagnostic and statistical manual of mental disorders (4th ed. rev). Washington, DC: American Psychiatric Association, 2000.

3. Mott JA, Wolfe MI, Alverson CJ, et al. National vehicle emissions policies and practices and declining US carbon monoxide-related mortality. JAMA 2002;288:988-95.

4. Thorpe M. Chronic carbon monoxide poisoning. Can J Psychiatry 1994;39:59-61.

5. Knobeloch L, Jackson R. Recognition of chronic carbon monoxide poisoning. WMJ 1999;98(6):26-9.

6. Turner M, Hamilton-Farrell MR, Clark RJ. Carbon monoxide poisoning: an update. J Accid Emerg Med 1999;16:92-6.

7. Unintentional carbon monoxide poisoning following winter storm—Washington January 1993. MMWR. 1993;42:109-11.

8. Wright J. Chronic and occult carbon monoxide poisoning: we don’t know what we’re missing. Emer Med J 2002;19:386-90.

9. Wald N, Idle M, Smith PG. Carboxyhaemoglobin levels in smokers of filter and plain cigarettes. Lancet 1977;1:110-12.

10. Raub JA, Benignus VA. Carbon monoxide and the nervous system. Neurosci Biobehav Rev 2002;26:925-40.

11. Ryan CM. Memory disturbances following chronic, low-level carbon monoxide exposure. Arch Clin Neuropsychol 1990;5:59-67.

12. Webb CJ, 2nd, Vaitkevicius PV. Dementia with a seasonal onset secondary to carbon monoxide poisoning. J Am Geriatr Soc 1997;45:1281-2.

13. Farrow JR, Davis GJ, Roy TM, et al. Fetal death due to nonlethal maternal carbon monoxide poisoning. J Forens Sci 1990;35:1448-52.

14. Vreman HJ, Mahoney JJ, Stevenson DK. Carbon monoxide and carboxyhemoglobin. Adv Pediatr 1995;42:303-34.

Issue
Current Psychiatry - 03(12)
Issue
Current Psychiatry - 03(12)
Page Number
79-88
Page Number
79-88
Publications
Publications
Article Type
Display Headline
Something in the air
Display Headline
Something in the air
Sections
Article Source

PURLs Copyright

Inside the Article

Commentary: Why patients may not respond to usual recommended dosages

Article Type
Changed
Mon, 04/16/2018 - 14:16
Display Headline
Commentary: Why patients may not respond to usual recommended dosages

Psychiatrists may consider using higher than usually recommended dosages of antipsychotics when faced with nonresponse. In this issue , Pierre et al1 carefully and thoughtfully discuss the pros and cons of this practice in patients with schizophrenia. Having reviewed that article, I thought CURRENT PSYCHIATRY’s readers might benefit from a theoretical framework for analyzing drug response.

‘Usual’ vs ‘unusual’ patients

A clinical trial for drug registration is, in essence, a population pharmacokinetic study whose goal is to determine the usual dosage for the usual patient in the trial. Many patients seen in clinical practice, such as those with treatment-refractory psychotic disorders, are typically excluded from registration trials. Thus, the usual registration trial patient may be an unusual patient in a clinician’s practice, and the trial’s usual dosage may not produce an adequate response for the clinician’s usual patient. How, then, might a clinician approach inadequate response, except by:

 

  • blindly exceeding the usually recommended dosage
  • switching among available drugs
  • adding drugs to create a complex cocktail?

This commentary dissects why a patient might not benefit from the usual recommended dosage and how that could lead to different courses of action.

Equation 1. Three variables (Table) determine response to any drug:

 

  • affinity for and intrinsic action on a regulatory protein (such as a receptor)
  • concentration (amount of drug reaching the site of action)
  • biological variance, which can shift an individual’s dose-response curve relative to that of the “usual” patient, making that individual more or less sensitive to the drug’s effects.2

Table

3 variables that determine patient response to any drug

 

Equation 1
Effect=Affinity for and intrinsic activity at a site of actionXDrug concentration (see Equation 2) Absorption Distribution Metabolism Elimination (ADME)XBiological variance Genetics Age Disease Environment (internal) (GADE)
Equation 2
Drug concentration = dosing rate/clearance

Equation 2. Drug concentration is dosing rate divided by clearance in a given patient. Dosing rate and clearance are equally important in determining drug concentration—which, in turn, determines the site of action engaged, to what degree, and the patient’s response to the drug.

Causes of inadequate response. Nonadherence is a common cause of inadequate response. When a patient repeatedly misses doses or stops taking the drug, the true dosing rate is lower than the prescribed dosing rate, resulting in reduced drug concentration and effect.

Pierre and colleagues focus on the “unusual” patient who does not respond optimally to antipsychotic dosages established in registration trials.1 As in Equation 1, sources of biological variance—genetics, age, disease, and environment (internal)—may distinguish the treatment-refractory patient from the responsive patient. The mnemonic GADE captures these variables:

Genetic variation refers to mutations in regulatory proteins that:

 

  • determine the drug’s action (such mutations may change the drug’s binding affinity, so that a higher concentration is needed to adequately engage the site)
  • determine what drug concentration reaches the site of action (such as drug-metabolizing enzymes that regulate clearance, or transporter proteins that prevent or facilitate the drug’s ability to reach the site of action).

Age refers to physiologic changes (pharmacodynamic or pharmacokinetic) that make the patient more or less sensitive to the drug’s effects.

Disease refers to differences in organ function related to pathophysiology. Patients with the same clinical presentation (in this case, psychosis) may respond differently to the same drug because they have different underlying pathophysiologies (such as schizophrenic syndrome due to differing genetic causes or to toxins or slow viruses).

Environment (internal) refers to exogenous substances in the body—such as drugs and dietary substances—that can interact with and influence response to other drugs.

Nonpsychiatric disease also can alter response to medication. For example, impaired hepatic, renal, or cardiac function can impair drug clearance, leading to greater-than-usual accumulation. Such a patient can be “sensitive” to the drug and experience a greater effect than is usually seen with the dosage given.

Dosing for clinical effect

Psychiatrists commonly titrate dosages based on clinical assessment of response.3 The clinician increases the dosage if a patient does not improve and has no obvious rate-limiting adverse effects.

Perhaps without realizing it, the clinician is assuming that the dosage is inadequate for a given patient because the concentration is inadequate due to rapid clearance. Other reasons are possible, however, such as:

 

  • the drug is not reaching the site of action a mutation at the site of action is altering
  • the drug’s binding affinity
  • the concentration may be too high, but the resulting adverse effects resemble worsening of the disease being treated. For example, akathisia due to dopamine-2 receptor blockade can present as agitation, and the clinician may increase the dosage when it should be decreased.
 

 

In the first two instances, escalating the dosage may be beneficial or cause toxicity. High levels in a peripheral compartment can cause adverse effects that may be silent until they become deadly (such as torsades de pointes). In the third instance, dosage escalation is the wrong step because the level is already too high.

Recommended dosage range

Principal goals of phase I studies in drug development are to establish the optimal dosage range and a maximum tolerated dosage. This upper limit is rarely, if ever, exceeded in later trials. Because phase I trial results are rarely published, the prescriber often does not know the rationale for a recommended dosing range’s upper limit.

Clinicians who escalate a drug’s dosage above the recommended range are using an n=1 paradigm, in which the patient is his or her own control. Unfortunately, treating one patient at a time cannot detect infrequent (much less rare) adverse events.

Using higher-than-recommended dosages thus exposes patients to unknown risks, with less monitoring than in a typical phase I trial in which subjects are confined to a research unit before, during, and after drug exposure. During the study, participants undergo serial ECGs, laboratory tests, and plasma drug level monitoring.

Therapeutic drug monitoring (TDM) is based on the concept that a meaningful relationship exists between a drug’s plasma concentration and its concentration at the site of action. Clinicians can measure the drug’s plasma concentration relative to the presumed dosage a patient is taking.

When nonadherence is the reason for nonresponse to usual dosing, TDM measurements of drug concentration would be lower than expected—or nonexistent with complete nonadherence. Rapid clearance, however, can also cause lower-than-expected levels on a given dosage.

So, how can the clinician determine whether the problem is rapid clearance or noncompliance? One way is to repeat the plasma level after arranging for supervised dosing for at least five times the half-life of the drug being measured. A higher level on follow-up would indicate that noncompliance is the likely problem. If the repeat level remains low, then the problem is most likely rapid clearance. In the latter case, the patient would need a higher dosage to achieve the concentrations associated with response in clinical trials.

Although TDM’s results are often conceptualized as being relative to a therapeutic range, TDM is fundamentally a means of measuring a patient’s ability to clear the drug. If the dosing rate and plasma drug level are known, then the clinician can solve for clearance by rearranging Equation 2. Rather than formally solving for clearance, results can be considered as within, below, or above the expected range for the dosage given. The clinician can then adjust the dosage to compensate for clearance that is faster or slower than usual. Thus, TDM allows clinicians to individualize dosages, taking into account the biological variances (Equation 1) that affect a patient’s ability to clear a specific drug.

TDM has limitations. It cannot assess whether a genetic mutation may be altering a drug’s binding affinity at the receptor site or whether the drug is not reaching the target compartment because of an abnormality in distribution mechanisms. Those possibilities would need to be assessed by techniques not available to most clinicians today.

Many psychiatrists think the inability to show a correlation between plasma drug levels and response is a limitation of TDM. That is not a limitation of TDM as much as a reflection on clinical trials of psychiatric drugs. Many such trials fail because of poor “signal-to-noise ratio”—defined as the true specific response to the treatment versus either placebo response or nonresponse due to not having an illness that is responsive to the drug.

Consider instead that the usually effective dosage defines a usually expected plasma drug concentration range associated with response. Further discussion of this topic is beyond the scope of this commentary, but the interested reader is referred to articles at www.preskorn.com, including Clinical pharmacology of serotonin selective re-uptake inhibitors (chapter 5); the column, Understanding dose-response curves in psychiatry; and the discussion, Finding the signal through the noise.

Summary

Based on the review by Pierre et al, the evidence for high-dose atypical antipsychotics’ safety and tolerability is not encouraging. These authors found only a modest body of evidence, and most study designs were not rigorous enough to eliminate erroneous conclusions. My intent here is not to advocate the use of higher-than-recommended dosages but to explain reasons why the patient may not respond and to call for more research.

Investigators designing future studies of nonresponse could consider including procedures to first rule out noncompliance and then divide participants into two groups:

 

 

 

  • patients who achieved usual plasma drug levels on the usual recommended dosages (normal clearance)
  • those who achieved levels below the usual expected range, despite good compliance (rapid clearance).

These two groups could then be randomized to continued exposure to the usual dosing range or higher-than-usual dosing. Patients with rapid clearance would be predicted to have a greater response to higher-than-usual dosing, compared with those with usual clearance.

In the absence of such trials, the clinician should proceed cautiously—if at all—to use higher-than usual antipsychotic dosages in his or her patients. The prescriber must always consider whether the risks outweigh the potential benefits, taking into account:

 

  • the drug’s therapeutic index
  • evidence of safety and tolerability problems in the individual patient as the dosage is escalated.

Related resources

 

  • Preskorn SH. The recommended dosage range: How is it established and why would it ever be exceeded? J Psychiatry Pract 2004;10(4):249-54.
References

 

1. Pierre JM, Wirshing DA, Wirshing WC. High-dose antipsychotic therapy: Desperation or data-driven. Current Psychiatry 2004;3(8):30-7.

2. Preskorn SH. Relating clinical trials to psychiatric practice: part I: the case of a 13-year-old on aripiprazole and fluoxetine. J Psychiatr Pract 2003;9(4):307-13. Also available at www.preskorn.com under Columns, Case studies.

3. Preskorn SH. Relating clinical trials to psychiatric practice: part II: the gap between the usual patient in registration trials and in practice. J Psychiatr Pract 2003;9(6):455-61. Also available at www.preskorn.com under Columns, Case studies.

Article PDF
Author and Disclosure Information

 

Sheldon H. Preskorn, MD
Professor and Chair, department of psychiatry University of Kansas School of Medicine, Wichita Associate Editor, CURRENT PSYCHIATRY

Sheldon H. Preskorn, MD Dr. Preskorn’s Web site at www.preskorn.com offers more information on this topic.

Issue
Current Psychiatry - 03(08)
Publications
Topics
Page Number
38-43
Sections
Author and Disclosure Information

 

Sheldon H. Preskorn, MD
Professor and Chair, department of psychiatry University of Kansas School of Medicine, Wichita Associate Editor, CURRENT PSYCHIATRY

Sheldon H. Preskorn, MD Dr. Preskorn’s Web site at www.preskorn.com offers more information on this topic.

Author and Disclosure Information

 

Sheldon H. Preskorn, MD
Professor and Chair, department of psychiatry University of Kansas School of Medicine, Wichita Associate Editor, CURRENT PSYCHIATRY

Sheldon H. Preskorn, MD Dr. Preskorn’s Web site at www.preskorn.com offers more information on this topic.

Article PDF
Article PDF

Psychiatrists may consider using higher than usually recommended dosages of antipsychotics when faced with nonresponse. In this issue , Pierre et al1 carefully and thoughtfully discuss the pros and cons of this practice in patients with schizophrenia. Having reviewed that article, I thought CURRENT PSYCHIATRY’s readers might benefit from a theoretical framework for analyzing drug response.

‘Usual’ vs ‘unusual’ patients

A clinical trial for drug registration is, in essence, a population pharmacokinetic study whose goal is to determine the usual dosage for the usual patient in the trial. Many patients seen in clinical practice, such as those with treatment-refractory psychotic disorders, are typically excluded from registration trials. Thus, the usual registration trial patient may be an unusual patient in a clinician’s practice, and the trial’s usual dosage may not produce an adequate response for the clinician’s usual patient. How, then, might a clinician approach inadequate response, except by:

 

  • blindly exceeding the usually recommended dosage
  • switching among available drugs
  • adding drugs to create a complex cocktail?

This commentary dissects why a patient might not benefit from the usual recommended dosage and how that could lead to different courses of action.

Equation 1. Three variables (Table) determine response to any drug:

 

  • affinity for and intrinsic action on a regulatory protein (such as a receptor)
  • concentration (amount of drug reaching the site of action)
  • biological variance, which can shift an individual’s dose-response curve relative to that of the “usual” patient, making that individual more or less sensitive to the drug’s effects.2

Table

3 variables that determine patient response to any drug

 

Equation 1
Effect=Affinity for and intrinsic activity at a site of actionXDrug concentration (see Equation 2) Absorption Distribution Metabolism Elimination (ADME)XBiological variance Genetics Age Disease Environment (internal) (GADE)
Equation 2
Drug concentration = dosing rate/clearance

Equation 2. Drug concentration is dosing rate divided by clearance in a given patient. Dosing rate and clearance are equally important in determining drug concentration—which, in turn, determines the site of action engaged, to what degree, and the patient’s response to the drug.

Causes of inadequate response. Nonadherence is a common cause of inadequate response. When a patient repeatedly misses doses or stops taking the drug, the true dosing rate is lower than the prescribed dosing rate, resulting in reduced drug concentration and effect.

Pierre and colleagues focus on the “unusual” patient who does not respond optimally to antipsychotic dosages established in registration trials.1 As in Equation 1, sources of biological variance—genetics, age, disease, and environment (internal)—may distinguish the treatment-refractory patient from the responsive patient. The mnemonic GADE captures these variables:

Genetic variation refers to mutations in regulatory proteins that:

 

  • determine the drug’s action (such mutations may change the drug’s binding affinity, so that a higher concentration is needed to adequately engage the site)
  • determine what drug concentration reaches the site of action (such as drug-metabolizing enzymes that regulate clearance, or transporter proteins that prevent or facilitate the drug’s ability to reach the site of action).

Age refers to physiologic changes (pharmacodynamic or pharmacokinetic) that make the patient more or less sensitive to the drug’s effects.

Disease refers to differences in organ function related to pathophysiology. Patients with the same clinical presentation (in this case, psychosis) may respond differently to the same drug because they have different underlying pathophysiologies (such as schizophrenic syndrome due to differing genetic causes or to toxins or slow viruses).

Environment (internal) refers to exogenous substances in the body—such as drugs and dietary substances—that can interact with and influence response to other drugs.

Nonpsychiatric disease also can alter response to medication. For example, impaired hepatic, renal, or cardiac function can impair drug clearance, leading to greater-than-usual accumulation. Such a patient can be “sensitive” to the drug and experience a greater effect than is usually seen with the dosage given.

Dosing for clinical effect

Psychiatrists commonly titrate dosages based on clinical assessment of response.3 The clinician increases the dosage if a patient does not improve and has no obvious rate-limiting adverse effects.

Perhaps without realizing it, the clinician is assuming that the dosage is inadequate for a given patient because the concentration is inadequate due to rapid clearance. Other reasons are possible, however, such as:

 

  • the drug is not reaching the site of action a mutation at the site of action is altering
  • the drug’s binding affinity
  • the concentration may be too high, but the resulting adverse effects resemble worsening of the disease being treated. For example, akathisia due to dopamine-2 receptor blockade can present as agitation, and the clinician may increase the dosage when it should be decreased.
 

 

In the first two instances, escalating the dosage may be beneficial or cause toxicity. High levels in a peripheral compartment can cause adverse effects that may be silent until they become deadly (such as torsades de pointes). In the third instance, dosage escalation is the wrong step because the level is already too high.

Recommended dosage range

Principal goals of phase I studies in drug development are to establish the optimal dosage range and a maximum tolerated dosage. This upper limit is rarely, if ever, exceeded in later trials. Because phase I trial results are rarely published, the prescriber often does not know the rationale for a recommended dosing range’s upper limit.

Clinicians who escalate a drug’s dosage above the recommended range are using an n=1 paradigm, in which the patient is his or her own control. Unfortunately, treating one patient at a time cannot detect infrequent (much less rare) adverse events.

Using higher-than-recommended dosages thus exposes patients to unknown risks, with less monitoring than in a typical phase I trial in which subjects are confined to a research unit before, during, and after drug exposure. During the study, participants undergo serial ECGs, laboratory tests, and plasma drug level monitoring.

Therapeutic drug monitoring (TDM) is based on the concept that a meaningful relationship exists between a drug’s plasma concentration and its concentration at the site of action. Clinicians can measure the drug’s plasma concentration relative to the presumed dosage a patient is taking.

When nonadherence is the reason for nonresponse to usual dosing, TDM measurements of drug concentration would be lower than expected—or nonexistent with complete nonadherence. Rapid clearance, however, can also cause lower-than-expected levels on a given dosage.

So, how can the clinician determine whether the problem is rapid clearance or noncompliance? One way is to repeat the plasma level after arranging for supervised dosing for at least five times the half-life of the drug being measured. A higher level on follow-up would indicate that noncompliance is the likely problem. If the repeat level remains low, then the problem is most likely rapid clearance. In the latter case, the patient would need a higher dosage to achieve the concentrations associated with response in clinical trials.

Although TDM’s results are often conceptualized as being relative to a therapeutic range, TDM is fundamentally a means of measuring a patient’s ability to clear the drug. If the dosing rate and plasma drug level are known, then the clinician can solve for clearance by rearranging Equation 2. Rather than formally solving for clearance, results can be considered as within, below, or above the expected range for the dosage given. The clinician can then adjust the dosage to compensate for clearance that is faster or slower than usual. Thus, TDM allows clinicians to individualize dosages, taking into account the biological variances (Equation 1) that affect a patient’s ability to clear a specific drug.

TDM has limitations. It cannot assess whether a genetic mutation may be altering a drug’s binding affinity at the receptor site or whether the drug is not reaching the target compartment because of an abnormality in distribution mechanisms. Those possibilities would need to be assessed by techniques not available to most clinicians today.

Many psychiatrists think the inability to show a correlation between plasma drug levels and response is a limitation of TDM. That is not a limitation of TDM as much as a reflection on clinical trials of psychiatric drugs. Many such trials fail because of poor “signal-to-noise ratio”—defined as the true specific response to the treatment versus either placebo response or nonresponse due to not having an illness that is responsive to the drug.

Consider instead that the usually effective dosage defines a usually expected plasma drug concentration range associated with response. Further discussion of this topic is beyond the scope of this commentary, but the interested reader is referred to articles at www.preskorn.com, including Clinical pharmacology of serotonin selective re-uptake inhibitors (chapter 5); the column, Understanding dose-response curves in psychiatry; and the discussion, Finding the signal through the noise.

Summary

Based on the review by Pierre et al, the evidence for high-dose atypical antipsychotics’ safety and tolerability is not encouraging. These authors found only a modest body of evidence, and most study designs were not rigorous enough to eliminate erroneous conclusions. My intent here is not to advocate the use of higher-than-recommended dosages but to explain reasons why the patient may not respond and to call for more research.

Investigators designing future studies of nonresponse could consider including procedures to first rule out noncompliance and then divide participants into two groups:

 

 

 

  • patients who achieved usual plasma drug levels on the usual recommended dosages (normal clearance)
  • those who achieved levels below the usual expected range, despite good compliance (rapid clearance).

These two groups could then be randomized to continued exposure to the usual dosing range or higher-than-usual dosing. Patients with rapid clearance would be predicted to have a greater response to higher-than-usual dosing, compared with those with usual clearance.

In the absence of such trials, the clinician should proceed cautiously—if at all—to use higher-than usual antipsychotic dosages in his or her patients. The prescriber must always consider whether the risks outweigh the potential benefits, taking into account:

 

  • the drug’s therapeutic index
  • evidence of safety and tolerability problems in the individual patient as the dosage is escalated.

Related resources

 

  • Preskorn SH. The recommended dosage range: How is it established and why would it ever be exceeded? J Psychiatry Pract 2004;10(4):249-54.

Psychiatrists may consider using higher than usually recommended dosages of antipsychotics when faced with nonresponse. In this issue , Pierre et al1 carefully and thoughtfully discuss the pros and cons of this practice in patients with schizophrenia. Having reviewed that article, I thought CURRENT PSYCHIATRY’s readers might benefit from a theoretical framework for analyzing drug response.

‘Usual’ vs ‘unusual’ patients

A clinical trial for drug registration is, in essence, a population pharmacokinetic study whose goal is to determine the usual dosage for the usual patient in the trial. Many patients seen in clinical practice, such as those with treatment-refractory psychotic disorders, are typically excluded from registration trials. Thus, the usual registration trial patient may be an unusual patient in a clinician’s practice, and the trial’s usual dosage may not produce an adequate response for the clinician’s usual patient. How, then, might a clinician approach inadequate response, except by:

 

  • blindly exceeding the usually recommended dosage
  • switching among available drugs
  • adding drugs to create a complex cocktail?

This commentary dissects why a patient might not benefit from the usual recommended dosage and how that could lead to different courses of action.

Equation 1. Three variables (Table) determine response to any drug:

 

  • affinity for and intrinsic action on a regulatory protein (such as a receptor)
  • concentration (amount of drug reaching the site of action)
  • biological variance, which can shift an individual’s dose-response curve relative to that of the “usual” patient, making that individual more or less sensitive to the drug’s effects.2

Table

3 variables that determine patient response to any drug

 

Equation 1
Effect=Affinity for and intrinsic activity at a site of actionXDrug concentration (see Equation 2) Absorption Distribution Metabolism Elimination (ADME)XBiological variance Genetics Age Disease Environment (internal) (GADE)
Equation 2
Drug concentration = dosing rate/clearance

Equation 2. Drug concentration is dosing rate divided by clearance in a given patient. Dosing rate and clearance are equally important in determining drug concentration—which, in turn, determines the site of action engaged, to what degree, and the patient’s response to the drug.

Causes of inadequate response. Nonadherence is a common cause of inadequate response. When a patient repeatedly misses doses or stops taking the drug, the true dosing rate is lower than the prescribed dosing rate, resulting in reduced drug concentration and effect.

Pierre and colleagues focus on the “unusual” patient who does not respond optimally to antipsychotic dosages established in registration trials.1 As in Equation 1, sources of biological variance—genetics, age, disease, and environment (internal)—may distinguish the treatment-refractory patient from the responsive patient. The mnemonic GADE captures these variables:

Genetic variation refers to mutations in regulatory proteins that:

 

  • determine the drug’s action (such mutations may change the drug’s binding affinity, so that a higher concentration is needed to adequately engage the site)
  • determine what drug concentration reaches the site of action (such as drug-metabolizing enzymes that regulate clearance, or transporter proteins that prevent or facilitate the drug’s ability to reach the site of action).

Age refers to physiologic changes (pharmacodynamic or pharmacokinetic) that make the patient more or less sensitive to the drug’s effects.

Disease refers to differences in organ function related to pathophysiology. Patients with the same clinical presentation (in this case, psychosis) may respond differently to the same drug because they have different underlying pathophysiologies (such as schizophrenic syndrome due to differing genetic causes or to toxins or slow viruses).

Environment (internal) refers to exogenous substances in the body—such as drugs and dietary substances—that can interact with and influence response to other drugs.

Nonpsychiatric disease also can alter response to medication. For example, impaired hepatic, renal, or cardiac function can impair drug clearance, leading to greater-than-usual accumulation. Such a patient can be “sensitive” to the drug and experience a greater effect than is usually seen with the dosage given.

Dosing for clinical effect

Psychiatrists commonly titrate dosages based on clinical assessment of response.3 The clinician increases the dosage if a patient does not improve and has no obvious rate-limiting adverse effects.

Perhaps without realizing it, the clinician is assuming that the dosage is inadequate for a given patient because the concentration is inadequate due to rapid clearance. Other reasons are possible, however, such as:

 

  • the drug is not reaching the site of action a mutation at the site of action is altering
  • the drug’s binding affinity
  • the concentration may be too high, but the resulting adverse effects resemble worsening of the disease being treated. For example, akathisia due to dopamine-2 receptor blockade can present as agitation, and the clinician may increase the dosage when it should be decreased.
 

 

In the first two instances, escalating the dosage may be beneficial or cause toxicity. High levels in a peripheral compartment can cause adverse effects that may be silent until they become deadly (such as torsades de pointes). In the third instance, dosage escalation is the wrong step because the level is already too high.

Recommended dosage range

Principal goals of phase I studies in drug development are to establish the optimal dosage range and a maximum tolerated dosage. This upper limit is rarely, if ever, exceeded in later trials. Because phase I trial results are rarely published, the prescriber often does not know the rationale for a recommended dosing range’s upper limit.

Clinicians who escalate a drug’s dosage above the recommended range are using an n=1 paradigm, in which the patient is his or her own control. Unfortunately, treating one patient at a time cannot detect infrequent (much less rare) adverse events.

Using higher-than-recommended dosages thus exposes patients to unknown risks, with less monitoring than in a typical phase I trial in which subjects are confined to a research unit before, during, and after drug exposure. During the study, participants undergo serial ECGs, laboratory tests, and plasma drug level monitoring.

Therapeutic drug monitoring (TDM) is based on the concept that a meaningful relationship exists between a drug’s plasma concentration and its concentration at the site of action. Clinicians can measure the drug’s plasma concentration relative to the presumed dosage a patient is taking.

When nonadherence is the reason for nonresponse to usual dosing, TDM measurements of drug concentration would be lower than expected—or nonexistent with complete nonadherence. Rapid clearance, however, can also cause lower-than-expected levels on a given dosage.

So, how can the clinician determine whether the problem is rapid clearance or noncompliance? One way is to repeat the plasma level after arranging for supervised dosing for at least five times the half-life of the drug being measured. A higher level on follow-up would indicate that noncompliance is the likely problem. If the repeat level remains low, then the problem is most likely rapid clearance. In the latter case, the patient would need a higher dosage to achieve the concentrations associated with response in clinical trials.

Although TDM’s results are often conceptualized as being relative to a therapeutic range, TDM is fundamentally a means of measuring a patient’s ability to clear the drug. If the dosing rate and plasma drug level are known, then the clinician can solve for clearance by rearranging Equation 2. Rather than formally solving for clearance, results can be considered as within, below, or above the expected range for the dosage given. The clinician can then adjust the dosage to compensate for clearance that is faster or slower than usual. Thus, TDM allows clinicians to individualize dosages, taking into account the biological variances (Equation 1) that affect a patient’s ability to clear a specific drug.

TDM has limitations. It cannot assess whether a genetic mutation may be altering a drug’s binding affinity at the receptor site or whether the drug is not reaching the target compartment because of an abnormality in distribution mechanisms. Those possibilities would need to be assessed by techniques not available to most clinicians today.

Many psychiatrists think the inability to show a correlation between plasma drug levels and response is a limitation of TDM. That is not a limitation of TDM as much as a reflection on clinical trials of psychiatric drugs. Many such trials fail because of poor “signal-to-noise ratio”—defined as the true specific response to the treatment versus either placebo response or nonresponse due to not having an illness that is responsive to the drug.

Consider instead that the usually effective dosage defines a usually expected plasma drug concentration range associated with response. Further discussion of this topic is beyond the scope of this commentary, but the interested reader is referred to articles at www.preskorn.com, including Clinical pharmacology of serotonin selective re-uptake inhibitors (chapter 5); the column, Understanding dose-response curves in psychiatry; and the discussion, Finding the signal through the noise.

Summary

Based on the review by Pierre et al, the evidence for high-dose atypical antipsychotics’ safety and tolerability is not encouraging. These authors found only a modest body of evidence, and most study designs were not rigorous enough to eliminate erroneous conclusions. My intent here is not to advocate the use of higher-than-recommended dosages but to explain reasons why the patient may not respond and to call for more research.

Investigators designing future studies of nonresponse could consider including procedures to first rule out noncompliance and then divide participants into two groups:

 

 

 

  • patients who achieved usual plasma drug levels on the usual recommended dosages (normal clearance)
  • those who achieved levels below the usual expected range, despite good compliance (rapid clearance).

These two groups could then be randomized to continued exposure to the usual dosing range or higher-than-usual dosing. Patients with rapid clearance would be predicted to have a greater response to higher-than-usual dosing, compared with those with usual clearance.

In the absence of such trials, the clinician should proceed cautiously—if at all—to use higher-than usual antipsychotic dosages in his or her patients. The prescriber must always consider whether the risks outweigh the potential benefits, taking into account:

 

  • the drug’s therapeutic index
  • evidence of safety and tolerability problems in the individual patient as the dosage is escalated.

Related resources

 

  • Preskorn SH. The recommended dosage range: How is it established and why would it ever be exceeded? J Psychiatry Pract 2004;10(4):249-54.
References

 

1. Pierre JM, Wirshing DA, Wirshing WC. High-dose antipsychotic therapy: Desperation or data-driven. Current Psychiatry 2004;3(8):30-7.

2. Preskorn SH. Relating clinical trials to psychiatric practice: part I: the case of a 13-year-old on aripiprazole and fluoxetine. J Psychiatr Pract 2003;9(4):307-13. Also available at www.preskorn.com under Columns, Case studies.

3. Preskorn SH. Relating clinical trials to psychiatric practice: part II: the gap between the usual patient in registration trials and in practice. J Psychiatr Pract 2003;9(6):455-61. Also available at www.preskorn.com under Columns, Case studies.

References

 

1. Pierre JM, Wirshing DA, Wirshing WC. High-dose antipsychotic therapy: Desperation or data-driven. Current Psychiatry 2004;3(8):30-7.

2. Preskorn SH. Relating clinical trials to psychiatric practice: part I: the case of a 13-year-old on aripiprazole and fluoxetine. J Psychiatr Pract 2003;9(4):307-13. Also available at www.preskorn.com under Columns, Case studies.

3. Preskorn SH. Relating clinical trials to psychiatric practice: part II: the gap between the usual patient in registration trials and in practice. J Psychiatr Pract 2003;9(6):455-61. Also available at www.preskorn.com under Columns, Case studies.

Issue
Current Psychiatry - 03(08)
Issue
Current Psychiatry - 03(08)
Page Number
38-43
Page Number
38-43
Publications
Publications
Topics
Article Type
Display Headline
Commentary: Why patients may not respond to usual recommended dosages
Display Headline
Commentary: Why patients may not respond to usual recommended dosages
Sections
Article PDF Media

Managing polypharmacy: Walking the fine line between help and harm

Article Type
Changed
Tue, 12/11/2018 - 15:12
Display Headline
Managing polypharmacy: Walking the fine line between help and harm

“Do no harm” is the first rule of medicine, yet 106,000 Americans die each year from properly prescribed and correctly taken medications.1 In some cases, the cause of death is known and can be attributed to a drug-drug interaction. The likelihood of death or hospitalization is directly proportional to the number of medications a patient is taking, even after controlling for underlying diseases.2

In psychiatry, it is not unusual for us to prescribe more than one psychotropic agent to manage a patient’s symptoms:

  • Patients with affective and psychotic disorders are commonly prescribed combinations of antipsychotics, mood stabilizers, antidepressants (often from more than one class), anxiolytics, antihistamines, and anticholinergics.
  • Patients with posttraumatic stress disorder may take selective serotonin reuptake inhibitors, buspirone, trazodone, antipsychotics, mood stabilizers, benzodiazepines, beta blockers, and opiates.
  • Multiple-drug regimens are used in treating other medical and psychiatric disorders, including chronic pain, fibromyalgia, chronic fatigue syndrome, sleep disorders, and epilepsy.

The greater the number of drugs used, the greater the likelihood that adverse events are emerging and are being treated, sometimes while being mistaken for patient psychopathology. As a prescriber, you are in a unique position to recognize and prevent interactions that can occur when patients are treated with two or more medications. This article defines polypharmacy, describes its consequences, prevalence, and risk factors, and offers an eight-step strategy with two mnemonics to help you avoid adverse events when prescribing multiple-drug regimens.

Box 1

POLYPHARMACY: MANY DRUGS, MANY DEFINITIONS

Poly, from the Greek word polus (many, much) and pharmacy, from the Greek word pharmakon (drug, poison) literally means many drugs or, alternatively, much poison.3 The word polypharmacy first appeared in the medical literature in 1959 in the New England Journal of Medicine4 and in the psychiatric literature in 1969 in an article citing its incidence at a state mental hospital.5

Many definitions have been used to describe and define polypharmacy, both qualitatively and quantitatively. Monotherapy is drug treatment with one drug. Sometimes treatment with two drugs is referred to as co-pharmacy, while treatment with three or more drugs is referred to as polypharmacy.Minor polypharmacy refers to treatment with two to four drugs, while major polypharmacyrefers to treatment with five or more drugs.6

What is polypharmacy?

Many definitions have been used to describe polypharmacy (Box 1).3-6 The most common definition is the use of five or more drugs at the same time in the same patient.7 Although polypharmacy often has a pejorative connotation, using five or more drugs may be therapeutic or contratherapeutic.

Therapeutic polypharmacy occurs, for example, when expert panels or researchers in carefully controlled clinical trials recommend using multiple medications to treat specific diseases. For example, the five-drug combination of isoniazid, rifampin, ethambutol, pyrazinamide, and pyridoxine is therapeutic in initial tuberculosis treatment. More is better in this case because four antibiotics are needed to prevent the development of multiple drug-resistant Mycobacterium tuberculosis, and adding pyridoxine prevents isoniazid-induced neurotoxicity. This example illustrates two prescribing principles:

  • using multiple drugs can help achieve an intended therapeutic goal
  • adding one drug can prevent a known side effect of another drug.

Another example is the therapeutic management of congestive heart failure, in which five drug classes—an angiotensin-converting enzyme (ACE) inhibitor, a diuretic, a digitalis glycoside, a beta blocker, and an aldosterone antagonist—are used in various combinations. All play a role in improving cardiac function and reducing morbidity and mortality.

Using combination drug therapy can also generate cost benefits, such as by adding a drug to delay or inhibit the metabolism of an expensive principal drug. For example, adding diltiazem—a cytochrome P450 (CYP) 3A4 inhibitor—to cyclosporine—which is metabolized by CYP 3A4 enzymes—reduces the dosage of cyclosporine needed to achieve a desired serum level, thereby reducing the cost of this drug. (Some have abandoned this strategy because of cyclosporine’s narrow therapeutic index.)

Contratherapeutic polypharmacy occurs when a patient taking multiple drugs experiences an unexpected or unintended adverse outcome.

Settings for polypharmacy

Polypharmacy occurs in five principal prescribing situations:

  • treatment of symptoms
  • treatment of multiple illnesses
  • treatment of phasic illnesses, such as many affective, anxiety, seizure, and neurodegenerative disorders
  • preventing or treating adverse effects of other drugs
  • attempting to accelerate the onset of action or augment the effects of a preceding drug.

As described above, diseases such as tuberculosis and congestive heart failure, with well-understood causes and pathophysiologies, are often treated with multiple therapeutic drug combinations. However, the causes of many psychiatric disorders and syndromes are less well-understood, which makes prescribing drug combinations more difficult. It may be that treating less well-understood diseases is a risk factor for contratherapeutic polypharmacy.

 

 

Most individuals who are prescribed five or more drugs are taking unique drug combinations.8 These heterogeneous regimens represent “an uncontrolled experiment,” with effects that cannot be predicted from studies in the literature.9Tables 1, 2, and 3 describe how contratherapeutic polypharmacy may occur with combinations of any number of drugs, whether five or more by the classic definition or only two. For example, contratherapeutic polypharmacy may occur when a patient is given the mood-stabilizing drugs valproate and carbamazepine (CBZ) at the same time.10 Here is why this combination may be dangerous:

  • Carbamazepine is oxidized by arene oxidase to CBZ 10,11-epoxide, which is hydrolyzed by epoxide hydrolase to CBZ 10,11-dihydroxide. The metabolite CBZ 10,11-epoxide has both therapeutic and toxic effects.
  • In monotherapy, the ratio of carbamazepine to CBZ 10,11-epoxide is 10:1, with CBZ 10,11-epoxide having a shorter half-life than carbamazepine.
  • However, when carbamazepine and valproate are taken as co-pharmacy, valproate blocks the hydrolysis of CBZ 10,11-epoxide by inhibiting epoxide hydrolase, so that the ratio of carbamazepine to CBZ 10,11-epoxide becomes 2:1. Higher concentrations of the epoxide metabolite contribute to neurotoxicity.

Table 1

POLYPHARMACY WITH TWO OR MORE MEDICATIONS

DescriptionExample
Two or more drugs from the same drug categoryTwo nonsteroidal anti-inflammatory drugs (NSAIDs), two ACE inhibitors, or two phenothiazines
Use of multiple medications across therapeutic classesUse of multiple CNS medications, as in multiple antidepressants, antipsychotics, or anticonvulsants
An inappropriate or unnecessary medication is prescribed to a patient taking other medicationInappropriate prescription due to relative or absolute contraindications Inappropriate prescription due to weak or no indication
Prescription of an exceedingly high dose to a patient taking other medicationThe maximum recommended dose may be functionally exceeded to a serious degree if a drug with a narrow therapeutic index (e.g., amitriptyline) is combined with one that blocks its metabolism (e.g., fluoxetine)
Two or more drugs sharing similar toxicitiesAnticholinergic toxicity due to combining a low-potency phenothiazine antipsychotic and a tertiary amine tricyclic antidepressant

Other examples of potentially dangerous drug combinations include those associated with torsades de pointes, which may occur with certain combinations of antihistamines, antidepressants, antipsychotics, antivirals, antibacterials, antifungals, antiarrhythmics, and promotility agents.

Drug-drug interactions

In a drug-drug interaction, the presence of one drug alters the nature, magnitude, or duration of the effect of a given dose of another drug; the interaction may be either therapeutic or adverse, depending on the desired effect. A drug-drug interaction may be intended or unintended and is determined by pharmacokinetics and pharmacodynamics rather than by therapeutic class.

Most available drug information describes the effects of individual drugs used alone (monopharmacy). Information on how one drug interacts with another (co-pharmacy) is more difficult to come by. A recent literature search using broad criteria for drug-drug interactions uncovered 4,277 indexed articles. Another search, this time using narrow criteria, produced only 316 articles, suggesting that systematic studies regarding drug-drug interactions are few.

Table 2

HOW PHARMACODYNAMICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

MechanismExamples
One drug has a mechanism of action directly opposing the mechanism of action of a co-prescribed drugBromocriptine and prochlorperazine in treating a patient with parkinsonism and nausea
Levidopa/carbidopa and risperidone in treating a patient with parkinsonism and psychosis
Venlafaxine and atenolol in treating a patient with depression and hypertension
One drug has an action that increases the potential for an adverse event of a co-prescribed drugOrthostatic hypotension and syncope when an ACE inhibitor is added to a diuretic
Orthostatic hypotension and syncope when risperidone, because of its action as an alpha-1 adrenergic blocker, is added to a diuretic
Narcosis and respiratory failure when parenteral fentanyl is added to oral meperidine
Neurotoxicity (absence status epilepticus) when valproate is added to clonazepam in children with absence seizures

However, if you understand the pharmacodynamics and pharmacokinetics that rule co-pharmacy, then you can apply this knowledge to more complex drug-drug interactions involving contratherapeutic polypharmacy.

How drug effects are determined. The nature and magnitude of a drug’s effect are determined by its site of action and its binding affinity, concentration, and action at that site.11 This relationship can be represented by the formula:

effect = potency at the site of action × concentration at the site of action

Potency at the site of action is determined by the binding affinity for the drug and the degree to which the receptor is stimulated or blocked, thus activating or inhibiting transmembrane and intracellular messengers (pharmacodynamics). Concentration at the site of action is determined by absorption, metabolism, distribution, and elimination (pharmacokinetics). Thus, the above model can be represented mathematically by:

effect = pharmacodynamics × pharmacokinetics

These factors determine a drug’s usual effect in the usual patient on the usual dosage, which is the goal of most clinical trials. However, all patients are not “usual,” because of inter-individual differences due to genetics, gender, age, environment, social habits such as smoking, intercurrent diseases affecting organ function, and concomitant drug therapy. Thus, when we take these factors into account, the first mathematical equation becomes:

 

 

effect = potency at the site of action × concentration at site of action × inter-individual variance

In other words, the clinical response equals the drug’s potency at the site of action times the drug’s concentration at the site of action times the patient’s underlying biology. Likewise, when we consider variability among patients, the second equation becomes:

effect = pharmacodynamics × pharmacokinetics × inter-individual variance

Table 3

HOW PHARMACOKINETICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

Mechanism of interaction of two or more drugsTwo or more drugs interact where …Examples
One negatively affects the other’s absorption Use of tetracycline with substances containing calcium
One negatively affects the other’s distribution Amiodarone and quinidine, by inhibiting P-glycoprotein, reduce the volume of distribution and/or clearance of digoxin, doubling its serum level
One negatively affects the other’s metabolismOne negatively affects the other’s oxidative metabolism by inducing CYP enzyme activityCarbamazepine induces CYP 2C9 and CYP 3A4 activity, which stimulates warfarin biotransformation, decreases its half-life, and lowers its serum concentration
 One negatively affects the other’s oxidative metabolism by inhibiting CYP enzyme activityKetoconazole inhibits CYP 3A4 activity, which inhibits terfenadine metabolism, resulting in serum terfenadine levels 32 to 100 times normal
 One inhibits hydroxylation of the other’s toxic metabolites, inhibiting their clearanceCombination of carbamazepine and valproate
One negatively affects the other’s elimination Lithium plus hydrochlorothiazide or an NSAID (both impair lithium excretion)

This addition to the equation explains how inter-individual variability can shift the dose-response curve to produce a greater or lesser effect than that which would be expected in the “usual” patient taking the prescribed dosage.

Inter-individual variance. The metabolism of dextromethorphan illustrates the effect of inter-individual variance. After a single dose, about 93% of Caucasians develop relatively lower dextromethorphan:dextrophan ratios, and about 7% develop relatively higher ratios. This difference defines patients who are pharmacogenetically CYP 2D6 extensive metabolizers versus those who are not.

Similarly, drugs sometimes cause biological variance, which predisposes to a drug-drug interaction. For example, the literature is replete with case reports and case series reporting that a substantial CYP 2D6 inhibitor—such as fluoxetine—blocks the metabolism of drugs that are principally metabolized by CYP 2D6. If the drug being metabolized has a narrow therapeutic index—such as amitriptyline—the resultant increase in its serum level can cause serious cardio and neurotoxicity, including arrhythmias, delirium, seizures, coma, and death.12

In such cases, a CYP 2D6 inhibitor converts the phenotype from a CYP 2D6 extensive metabolizer into a CYP 2D6 poor metabolizer. Hence, the clinician must consider how a specific patient may differ from the usual patient when selecting and dosing a drug. The difference may be genetic or acquired, as in this example.

Table 4

RISK FACTORS FOR POLYPHARMACY

Psychiatric disordersMedications being taken
SchizophreniaCardiovascular agents
Bipolar disorderAntipsychotics
DepressionMood stabilizers
Borderline and other personality disordersAntidepressants
Substance abuse (including tobacco habituation)Self-medication with aspirin
Neurologic disordersDemographic variables
Mental retardationAge 65 or older
DementiaEthnicity (Caucasian, African-American)
Chronic pain, facial painFemale gender
Headache (including migraine)Psychosocial variables
InsomniaLower socioeconomic status
EpilepsyInner-city residence
Medical disordersLower level of education
Chronic diseases, multiple diseasesUnemployment
ObesitySelf-medication
DiabetesConcealed drug use
Chronic hypertension 
Coronary artery disease 

The following equation explains how dose is related to drug concentration, which takes into account the drug’s pharmacokinetics:

drug concentration = dosing rate (mg/day) ÷ clearance (ml/min)

In other words, the concentration achieved in a specific patient is determined by the dosage relative to the patient’s ability to clear the drug from the body.

Consequences, prevalence of polypharmacy

Polypharmacy increases patients’ risk for many ill effects, including incidence and severity of adverse events, drug-drug interactions, medication errors, hospitalizations, morbidity, mortality, and direct and indirect costs. At least 12 reports and studies have been published showing the association between polypharmacy and death,2,13-23 and in some of these reports the association is present even after controlling for underlying diseases.

The prevalence of polypharmacy varies by country and population. In Denmark, for example, the prevalence of polypharmacy is approximately 1.2%,6 compared with approximately 7% in the United States.24 Nearly one-half (46%) of all elderly persons admitted to U.S. hospitals may be taking seven or more medications.25 Polypharmacy is especially problematic in patients age 65 and older (Box 2),26-31 in whom the top five preventable threats to health are congestive heart failure, breast cancer, hypertension, pneumonia, and adverse drug events.32 Although older persons make up less than 15% of the population, they take the greatest number and quantity of medications, purchase 40% of all nonprescription medications, and use 33% of all retail prescriptions.30

Box 2

POLYPHARMACY RISKS IN PATIENTS AGE 65 AND OLDER

  • 14% of older patients prescribed psychotropics experience a hip fracture, accounting for 32,000 annual hip fractures in the United States.26
  • 28% of older patients’ hospitalizations are due to adverse events or non-adherence to drug therapy.27
  • 35% of older patients taking three or more prescription medications at hospital discharge are re-hospitalized within 6 months. Problems with medications lead to 6.4% of these re-admissions.28
  • Among older drivers, taking a psychoactive drug multiplies the risk of a motor vehicle accident involving injuries by 1.5 to 5.5 times. The greater the dosage, the greater the risk.29
  • Hospital admissions related to adverse events from medications in older patients cost $20 billion annually (excluding indirect costs).30
  • Morbidity and mortality related to drug therapy in ambulatory patients in the United States costs $76.6 billion annually.31
 

 

Psychiatric disorders including schizophrenia, bipolar disorder, depression, personality disorders, and substance abuse place patients at higher risk for polypharmacy, as do certain demographic, psychosocial, medication, medical, and neurologic factors (Table 4). Other factors that increase the risk for polypharmacy include:

  • institutional factors (recent hospitalization, admission to a surgical ward, nursing home placement, home health care, increased number of pharmacies used, increased number of clinics attended, client-centered psychiatric treatment compared with non-client-centered psychiatric treatment)
  • provider factors (visit to a physician, treatment by general practitioners compared with specialists, increased number of providers, undocumented rationale or diagnosis supporting multiple medication use)
  • having medical insurance.

Steps to avoiding polypharmacy

By identifying polypharmacy’s risk factors, we may decrease its associated morbidity, mortality, and cost. Steps to follow while prescribing—as represented by the mnemonics SAIL33 and TIDE—may help you avoid polypharmacy’s negative consequences.

SAIL. Keep the drug regimen as simple as possible. Aim for once-daily or twice daily dosing. Try to simplify complex drug regimens by discontinuing any drug that does not achieve its defined therapeutic goal. For diseases and syndromes with less clear-cut causes, subtracting drugs from a complicated regimen may be more therapeutic than adding another drug. Try to treat multiple symptoms and syndromes with a single drug that may have multiple beneficial effects, rather than treating each symptom or syndrome with individual drugs.

Understand the potential adverse effects of each drug and potential drug-drug interactions. Whenever practical, choose drugs with broad rather than narrow therapeutic indices.

Each prescribed drug should have a clear indication and a well-defined therapeutic goal. Prescribe using evidence-based medicine as much as is practical.

List the name and dosage of each drug in the patient’s chart, and provide this information to the patient.33 Consider adopting computer data entry and feedback procedures, which have been shown to decrease polypharmacy34 and drug-drug interactions.35

TIDE. In the busy medical practice, writing a prescription signals to the patient that his or her time with the doctor is almost finished. Allow time to address medication issues.

Apply the understanding of individual variability, pharmacokinetics, and pharmacodynamics when prescribing. Review with the patient all prescription and nonprescription drugs and dietary supplements being taken.

Be careful to avoid potentially dangerous drug-drug interactions, especially those associated with serious adverse events such as torsades de pointes.

Educate patients regarding drug and non-drug treatments. Explain potential adverse effects of each drug and potential drug-drug interactions.

Related resources

Disclosure

Drs. Werder and Preskorn have served on the speakers bureau of, as consultants to, or as principal investigators for Abbott Laboratories, AstraZeneca Pharmaceuticals, Biovail Corp., Bristol-Meyers Squibb Co., Merck and Co., Eisai Inc., Eli Lilly and Co., GlaxoSmithKline, Hoffman-LaRoche, Janssen Pharmaceutica, Lundbeck, Novartis Pharmaceuticals Corp., Organon, Pfizer Inc., Solvay, Wyeth Pharmaceuticals, and Yamanouchi Pharmaceuticals Co., Ltd.

References

1. Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 1998;279(15):1200-5.

2. Incalzi RA, Gemma A, Capparella O, et al. Predicting mortality and length of stay of geriatric patients in an acute care general hospital. J Gerontol 1992;47(2):M35-9.

3. Berube MS, Neely DJ, DeVinne PB. American Heritage Dictionary. (2nd College ed). Boston: Houghton Mifflin Co, 1982.

4. Friend DG. Polypharmacy: multiple-ingredient and shotgun prescriptions. N Engl J Med 1959;260(20):1015-8.

5. Sheppard C, Collins L, Fiorentino D, Fracchia J, Merlis S. Polypharmacy in psychiatric treatment. I. Incidence at a state hospital. Curr Ther Res Clin Exp 1969;(12):765-74.

6. Bjerrum L, Rosholm JU, Hallas J, Kragstrup J. Methods for estimating the occurrence of polypharmacy by means of a prescription database. Eur J Clin Pharmacol 1997;53(1):7-11.

7. Werder SF. Polypharmacy: definitions and risk factors (grand rounds). University of Kansas School of Medicine-Wichita, Department of Psychiatry and Behavioral Sciences. Via Christi Regional Medical Center, St. Joseph Campus: Dec 12, 2000.

8. Bjerrum L, Sogaard J, Hallas J, Kragstrup J. Polypharmacy: correlations with sex, age and drug regimen. A prescription database study. Eur J Clin Pharmacol 1998;54(3):197-202.

9. Bjerrum L. Pharmacoepidemiological Studies of Polypharmacy: Methodological issues, population estimates, and influence of practice patterns (PhD thesis). Odense University Faculty of Health Sciences, Department of clinical pharmacology and research unit of general practice. Denmark; 1998. Available at http://www.sdu.dk/health/IPH/genpract/staff/lbjerrum/PHD/PHD.HTM. Accessed Jan. 9, 2003.

10. Wilder BJ. Pharmacokinetics of valproate and carbamazepine. J Clin Psychopharmacol 1992;12(1 suppl):64S-68S.

11. Preskorn SH. The rational basis for the development and use of newer antidepressants. In: Outpatient management of depression: a guide for the practitioner (2nd ed). Caddo, OK: Professional Publications, Inc; 1999;57-103.

12. Vaughan DA. Interaction of fluoxetine with tricyclic antidepressants. Am J Psychiatry 1988;145(11):1478.-

13. Meeker JE, Reynolds PC. Postmortem tissue methamphetamine concentrations following selegiline administration. J Anal Toxicol 1990;14(5):330-1.

14. Sallee FR, DeVane CL, Ferrell RE. Fluoxetine-related death in a child with cytochrome P-450 2D6 genetic deficiency. J Child Adolesc Psychopharmacol 2000;10(1):27-34.

15. Ellis RJ, Mayo MS, Bodensteiner DM. Ciprofloxacin-warfarin coagulopathy: a case series. Am J Hematol 2000;63(1):28-31.

16. Konig SA, Siemes H, Blaker F, et al. Severe hepatotoxicity during valproate therapy: an update and report of eight new fatalities. Epilepsia 1994;35(5):1005-15.

17. Fattinger K, Roos M, Vergeres P, et al. Epidemiology of drug exposure and adverse drug reactions in two Swiss departments of internal medicine. Br J Clin Pharmacol 2000;49(2):158-67.

18. Ebbesen J, Buajordet I, Erikssen J, Svaar H, Brors O, Hilberg T. Drugs as a cause of death. A prospective quality assurance project in a department of medicine (Norwegian). Tidsskr Nor Laegeforen 1995;115(19):2369-72.

19. Alarcon T, Barcena A, Gonzalez-Montalvo JI, Penalosa C, Salgado A. Factors predictive of outcome on admission to an acute geriatric ward. Age Ageing 1999;28(5):429-32.

20. Smith NK, Albazzaz MK. A prospective study of urinary retention and risk of death after proximal femoral fracture. Age Ageing 1996;25(2):150-4.

21. Pulska T, Pahkala K, Laippala P, Kivela SL. Six-year survival of depressed elderly Finns: a community study. Int J Geriatr Psychiatry 1997;12(9):942-50.

22. Waddington JL, Youssef HA, Kinsella A. Mortality in schizophrenia. Antipsychotic polypharmacy and absence of adjunctive anticholinergics over the course of a 10-year prospective study. Br J Psychiatry 1998;173:325-9.

23. Burns R, Nichols LO, Graney MJ, Cloar FT. Impact of continued geriatric outpatient management on health outcomes of older veterans. Arch Intern Med 1995;155(12):1313-8.

24. Kaufman DW, Kelly JP, Rosenberg L, Anderson TE, Mitchell AA. Recent patterns of medication use in the ambulatory adult population of the United States: the Slone survey. JAMA 2002;16;287(3):337-44.

25. Flaherty JH, Perry HM, 3rd, Lynchard GS, Morley JE. Polypharmacy and hospitalization among older home care patients. J Gerontol A Biol Sci Med Sci 2000;55(10):M554-9.

26. Ray WA, Griffin MR, Schaffner W, Baugh DK, Melton LJ, 3rd. Psychotropic drug use and the risk of hip fracture. N Engl J Med 1987;316(7):363-9.

27. Col N, Fanale JE, Kronholm P. The role of medication noncompliance and adverse drug reactions in hospitalizations of the elderly. Arch Intern Med 1990;150(4):841-5.

28. Bero LA, Lipton HL, Bird JA. Characterization of geriatric drug-related hospital readmissions. Med Care 1991;29(10):989-1003.

29. Ray WA, Fought RL, Decker MD. Psychoactive drugs and the risk of injurious motor vehicle crashes in elderly drivers. Am J Epidemiol 1992;136(7):873-83.

30. Prescription drugs and the elderly. Publication AO/HEHS-95-152. Washington, DC: U.S. General Accounting Office, July 1995.

31. Johnson JA, Bootman JL. Drug-related morbidity and mortality. A cost-of-illness model. Arch Intern Med 1995;155(18):1949-56.

32. Fink A, Siu AL, Brook RH, Park RE, Solomon DH. Assuring the quality of health care for older persons. An expert panel’s priorities. JAMA 1987;258(14):1905-8.

33. Lee DR. Polypharmacy: a case report and new protocol for management. J Am Board Fam Pract 1998;11(2):140-4.

34. Hamdy RC, Moore SW, Whalen K, et al. Reducing polypharmacy in extended care. South Med J 1995;88(5):534-8.

35. Haumschild MJ, Ward ES, Bishop JM, Haumschild MS. Pharmacy-based computer system for monitoring and reporting drug interactions. Am J Hosp Pharm 1987;44(2):345-8.

Article PDF
Author and Disclosure Information

Steven F. Werder, DO
Assistant professor Departments of psychiatry and internal medicine University of Kansas School of Medicine Wichita

Sheldon H. Preskorn, MD
Professor and chairman Department of psychiatry and behavioral sciences

University of Kansas School of Medicine Wichita

Issue
Current Psychiatry - 02(02)
Publications
Topics
Page Number
24-36
Sections
Author and Disclosure Information

Steven F. Werder, DO
Assistant professor Departments of psychiatry and internal medicine University of Kansas School of Medicine Wichita

Sheldon H. Preskorn, MD
Professor and chairman Department of psychiatry and behavioral sciences

University of Kansas School of Medicine Wichita

Author and Disclosure Information

Steven F. Werder, DO
Assistant professor Departments of psychiatry and internal medicine University of Kansas School of Medicine Wichita

Sheldon H. Preskorn, MD
Professor and chairman Department of psychiatry and behavioral sciences

University of Kansas School of Medicine Wichita

Article PDF
Article PDF

“Do no harm” is the first rule of medicine, yet 106,000 Americans die each year from properly prescribed and correctly taken medications.1 In some cases, the cause of death is known and can be attributed to a drug-drug interaction. The likelihood of death or hospitalization is directly proportional to the number of medications a patient is taking, even after controlling for underlying diseases.2

In psychiatry, it is not unusual for us to prescribe more than one psychotropic agent to manage a patient’s symptoms:

  • Patients with affective and psychotic disorders are commonly prescribed combinations of antipsychotics, mood stabilizers, antidepressants (often from more than one class), anxiolytics, antihistamines, and anticholinergics.
  • Patients with posttraumatic stress disorder may take selective serotonin reuptake inhibitors, buspirone, trazodone, antipsychotics, mood stabilizers, benzodiazepines, beta blockers, and opiates.
  • Multiple-drug regimens are used in treating other medical and psychiatric disorders, including chronic pain, fibromyalgia, chronic fatigue syndrome, sleep disorders, and epilepsy.

The greater the number of drugs used, the greater the likelihood that adverse events are emerging and are being treated, sometimes while being mistaken for patient psychopathology. As a prescriber, you are in a unique position to recognize and prevent interactions that can occur when patients are treated with two or more medications. This article defines polypharmacy, describes its consequences, prevalence, and risk factors, and offers an eight-step strategy with two mnemonics to help you avoid adverse events when prescribing multiple-drug regimens.

Box 1

POLYPHARMACY: MANY DRUGS, MANY DEFINITIONS

Poly, from the Greek word polus (many, much) and pharmacy, from the Greek word pharmakon (drug, poison) literally means many drugs or, alternatively, much poison.3 The word polypharmacy first appeared in the medical literature in 1959 in the New England Journal of Medicine4 and in the psychiatric literature in 1969 in an article citing its incidence at a state mental hospital.5

Many definitions have been used to describe and define polypharmacy, both qualitatively and quantitatively. Monotherapy is drug treatment with one drug. Sometimes treatment with two drugs is referred to as co-pharmacy, while treatment with three or more drugs is referred to as polypharmacy.Minor polypharmacy refers to treatment with two to four drugs, while major polypharmacyrefers to treatment with five or more drugs.6

What is polypharmacy?

Many definitions have been used to describe polypharmacy (Box 1).3-6 The most common definition is the use of five or more drugs at the same time in the same patient.7 Although polypharmacy often has a pejorative connotation, using five or more drugs may be therapeutic or contratherapeutic.

Therapeutic polypharmacy occurs, for example, when expert panels or researchers in carefully controlled clinical trials recommend using multiple medications to treat specific diseases. For example, the five-drug combination of isoniazid, rifampin, ethambutol, pyrazinamide, and pyridoxine is therapeutic in initial tuberculosis treatment. More is better in this case because four antibiotics are needed to prevent the development of multiple drug-resistant Mycobacterium tuberculosis, and adding pyridoxine prevents isoniazid-induced neurotoxicity. This example illustrates two prescribing principles:

  • using multiple drugs can help achieve an intended therapeutic goal
  • adding one drug can prevent a known side effect of another drug.

Another example is the therapeutic management of congestive heart failure, in which five drug classes—an angiotensin-converting enzyme (ACE) inhibitor, a diuretic, a digitalis glycoside, a beta blocker, and an aldosterone antagonist—are used in various combinations. All play a role in improving cardiac function and reducing morbidity and mortality.

Using combination drug therapy can also generate cost benefits, such as by adding a drug to delay or inhibit the metabolism of an expensive principal drug. For example, adding diltiazem—a cytochrome P450 (CYP) 3A4 inhibitor—to cyclosporine—which is metabolized by CYP 3A4 enzymes—reduces the dosage of cyclosporine needed to achieve a desired serum level, thereby reducing the cost of this drug. (Some have abandoned this strategy because of cyclosporine’s narrow therapeutic index.)

Contratherapeutic polypharmacy occurs when a patient taking multiple drugs experiences an unexpected or unintended adverse outcome.

Settings for polypharmacy

Polypharmacy occurs in five principal prescribing situations:

  • treatment of symptoms
  • treatment of multiple illnesses
  • treatment of phasic illnesses, such as many affective, anxiety, seizure, and neurodegenerative disorders
  • preventing or treating adverse effects of other drugs
  • attempting to accelerate the onset of action or augment the effects of a preceding drug.

As described above, diseases such as tuberculosis and congestive heart failure, with well-understood causes and pathophysiologies, are often treated with multiple therapeutic drug combinations. However, the causes of many psychiatric disorders and syndromes are less well-understood, which makes prescribing drug combinations more difficult. It may be that treating less well-understood diseases is a risk factor for contratherapeutic polypharmacy.

 

 

Most individuals who are prescribed five or more drugs are taking unique drug combinations.8 These heterogeneous regimens represent “an uncontrolled experiment,” with effects that cannot be predicted from studies in the literature.9Tables 1, 2, and 3 describe how contratherapeutic polypharmacy may occur with combinations of any number of drugs, whether five or more by the classic definition or only two. For example, contratherapeutic polypharmacy may occur when a patient is given the mood-stabilizing drugs valproate and carbamazepine (CBZ) at the same time.10 Here is why this combination may be dangerous:

  • Carbamazepine is oxidized by arene oxidase to CBZ 10,11-epoxide, which is hydrolyzed by epoxide hydrolase to CBZ 10,11-dihydroxide. The metabolite CBZ 10,11-epoxide has both therapeutic and toxic effects.
  • In monotherapy, the ratio of carbamazepine to CBZ 10,11-epoxide is 10:1, with CBZ 10,11-epoxide having a shorter half-life than carbamazepine.
  • However, when carbamazepine and valproate are taken as co-pharmacy, valproate blocks the hydrolysis of CBZ 10,11-epoxide by inhibiting epoxide hydrolase, so that the ratio of carbamazepine to CBZ 10,11-epoxide becomes 2:1. Higher concentrations of the epoxide metabolite contribute to neurotoxicity.

Table 1

POLYPHARMACY WITH TWO OR MORE MEDICATIONS

DescriptionExample
Two or more drugs from the same drug categoryTwo nonsteroidal anti-inflammatory drugs (NSAIDs), two ACE inhibitors, or two phenothiazines
Use of multiple medications across therapeutic classesUse of multiple CNS medications, as in multiple antidepressants, antipsychotics, or anticonvulsants
An inappropriate or unnecessary medication is prescribed to a patient taking other medicationInappropriate prescription due to relative or absolute contraindications Inappropriate prescription due to weak or no indication
Prescription of an exceedingly high dose to a patient taking other medicationThe maximum recommended dose may be functionally exceeded to a serious degree if a drug with a narrow therapeutic index (e.g., amitriptyline) is combined with one that blocks its metabolism (e.g., fluoxetine)
Two or more drugs sharing similar toxicitiesAnticholinergic toxicity due to combining a low-potency phenothiazine antipsychotic and a tertiary amine tricyclic antidepressant

Other examples of potentially dangerous drug combinations include those associated with torsades de pointes, which may occur with certain combinations of antihistamines, antidepressants, antipsychotics, antivirals, antibacterials, antifungals, antiarrhythmics, and promotility agents.

Drug-drug interactions

In a drug-drug interaction, the presence of one drug alters the nature, magnitude, or duration of the effect of a given dose of another drug; the interaction may be either therapeutic or adverse, depending on the desired effect. A drug-drug interaction may be intended or unintended and is determined by pharmacokinetics and pharmacodynamics rather than by therapeutic class.

Most available drug information describes the effects of individual drugs used alone (monopharmacy). Information on how one drug interacts with another (co-pharmacy) is more difficult to come by. A recent literature search using broad criteria for drug-drug interactions uncovered 4,277 indexed articles. Another search, this time using narrow criteria, produced only 316 articles, suggesting that systematic studies regarding drug-drug interactions are few.

Table 2

HOW PHARMACODYNAMICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

MechanismExamples
One drug has a mechanism of action directly opposing the mechanism of action of a co-prescribed drugBromocriptine and prochlorperazine in treating a patient with parkinsonism and nausea
Levidopa/carbidopa and risperidone in treating a patient with parkinsonism and psychosis
Venlafaxine and atenolol in treating a patient with depression and hypertension
One drug has an action that increases the potential for an adverse event of a co-prescribed drugOrthostatic hypotension and syncope when an ACE inhibitor is added to a diuretic
Orthostatic hypotension and syncope when risperidone, because of its action as an alpha-1 adrenergic blocker, is added to a diuretic
Narcosis and respiratory failure when parenteral fentanyl is added to oral meperidine
Neurotoxicity (absence status epilepticus) when valproate is added to clonazepam in children with absence seizures

However, if you understand the pharmacodynamics and pharmacokinetics that rule co-pharmacy, then you can apply this knowledge to more complex drug-drug interactions involving contratherapeutic polypharmacy.

How drug effects are determined. The nature and magnitude of a drug’s effect are determined by its site of action and its binding affinity, concentration, and action at that site.11 This relationship can be represented by the formula:

effect = potency at the site of action × concentration at the site of action

Potency at the site of action is determined by the binding affinity for the drug and the degree to which the receptor is stimulated or blocked, thus activating or inhibiting transmembrane and intracellular messengers (pharmacodynamics). Concentration at the site of action is determined by absorption, metabolism, distribution, and elimination (pharmacokinetics). Thus, the above model can be represented mathematically by:

effect = pharmacodynamics × pharmacokinetics

These factors determine a drug’s usual effect in the usual patient on the usual dosage, which is the goal of most clinical trials. However, all patients are not “usual,” because of inter-individual differences due to genetics, gender, age, environment, social habits such as smoking, intercurrent diseases affecting organ function, and concomitant drug therapy. Thus, when we take these factors into account, the first mathematical equation becomes:

 

 

effect = potency at the site of action × concentration at site of action × inter-individual variance

In other words, the clinical response equals the drug’s potency at the site of action times the drug’s concentration at the site of action times the patient’s underlying biology. Likewise, when we consider variability among patients, the second equation becomes:

effect = pharmacodynamics × pharmacokinetics × inter-individual variance

Table 3

HOW PHARMACOKINETICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

Mechanism of interaction of two or more drugsTwo or more drugs interact where …Examples
One negatively affects the other’s absorption Use of tetracycline with substances containing calcium
One negatively affects the other’s distribution Amiodarone and quinidine, by inhibiting P-glycoprotein, reduce the volume of distribution and/or clearance of digoxin, doubling its serum level
One negatively affects the other’s metabolismOne negatively affects the other’s oxidative metabolism by inducing CYP enzyme activityCarbamazepine induces CYP 2C9 and CYP 3A4 activity, which stimulates warfarin biotransformation, decreases its half-life, and lowers its serum concentration
 One negatively affects the other’s oxidative metabolism by inhibiting CYP enzyme activityKetoconazole inhibits CYP 3A4 activity, which inhibits terfenadine metabolism, resulting in serum terfenadine levels 32 to 100 times normal
 One inhibits hydroxylation of the other’s toxic metabolites, inhibiting their clearanceCombination of carbamazepine and valproate
One negatively affects the other’s elimination Lithium plus hydrochlorothiazide or an NSAID (both impair lithium excretion)

This addition to the equation explains how inter-individual variability can shift the dose-response curve to produce a greater or lesser effect than that which would be expected in the “usual” patient taking the prescribed dosage.

Inter-individual variance. The metabolism of dextromethorphan illustrates the effect of inter-individual variance. After a single dose, about 93% of Caucasians develop relatively lower dextromethorphan:dextrophan ratios, and about 7% develop relatively higher ratios. This difference defines patients who are pharmacogenetically CYP 2D6 extensive metabolizers versus those who are not.

Similarly, drugs sometimes cause biological variance, which predisposes to a drug-drug interaction. For example, the literature is replete with case reports and case series reporting that a substantial CYP 2D6 inhibitor—such as fluoxetine—blocks the metabolism of drugs that are principally metabolized by CYP 2D6. If the drug being metabolized has a narrow therapeutic index—such as amitriptyline—the resultant increase in its serum level can cause serious cardio and neurotoxicity, including arrhythmias, delirium, seizures, coma, and death.12

In such cases, a CYP 2D6 inhibitor converts the phenotype from a CYP 2D6 extensive metabolizer into a CYP 2D6 poor metabolizer. Hence, the clinician must consider how a specific patient may differ from the usual patient when selecting and dosing a drug. The difference may be genetic or acquired, as in this example.

Table 4

RISK FACTORS FOR POLYPHARMACY

Psychiatric disordersMedications being taken
SchizophreniaCardiovascular agents
Bipolar disorderAntipsychotics
DepressionMood stabilizers
Borderline and other personality disordersAntidepressants
Substance abuse (including tobacco habituation)Self-medication with aspirin
Neurologic disordersDemographic variables
Mental retardationAge 65 or older
DementiaEthnicity (Caucasian, African-American)
Chronic pain, facial painFemale gender
Headache (including migraine)Psychosocial variables
InsomniaLower socioeconomic status
EpilepsyInner-city residence
Medical disordersLower level of education
Chronic diseases, multiple diseasesUnemployment
ObesitySelf-medication
DiabetesConcealed drug use
Chronic hypertension 
Coronary artery disease 

The following equation explains how dose is related to drug concentration, which takes into account the drug’s pharmacokinetics:

drug concentration = dosing rate (mg/day) ÷ clearance (ml/min)

In other words, the concentration achieved in a specific patient is determined by the dosage relative to the patient’s ability to clear the drug from the body.

Consequences, prevalence of polypharmacy

Polypharmacy increases patients’ risk for many ill effects, including incidence and severity of adverse events, drug-drug interactions, medication errors, hospitalizations, morbidity, mortality, and direct and indirect costs. At least 12 reports and studies have been published showing the association between polypharmacy and death,2,13-23 and in some of these reports the association is present even after controlling for underlying diseases.

The prevalence of polypharmacy varies by country and population. In Denmark, for example, the prevalence of polypharmacy is approximately 1.2%,6 compared with approximately 7% in the United States.24 Nearly one-half (46%) of all elderly persons admitted to U.S. hospitals may be taking seven or more medications.25 Polypharmacy is especially problematic in patients age 65 and older (Box 2),26-31 in whom the top five preventable threats to health are congestive heart failure, breast cancer, hypertension, pneumonia, and adverse drug events.32 Although older persons make up less than 15% of the population, they take the greatest number and quantity of medications, purchase 40% of all nonprescription medications, and use 33% of all retail prescriptions.30

Box 2

POLYPHARMACY RISKS IN PATIENTS AGE 65 AND OLDER

  • 14% of older patients prescribed psychotropics experience a hip fracture, accounting for 32,000 annual hip fractures in the United States.26
  • 28% of older patients’ hospitalizations are due to adverse events or non-adherence to drug therapy.27
  • 35% of older patients taking three or more prescription medications at hospital discharge are re-hospitalized within 6 months. Problems with medications lead to 6.4% of these re-admissions.28
  • Among older drivers, taking a psychoactive drug multiplies the risk of a motor vehicle accident involving injuries by 1.5 to 5.5 times. The greater the dosage, the greater the risk.29
  • Hospital admissions related to adverse events from medications in older patients cost $20 billion annually (excluding indirect costs).30
  • Morbidity and mortality related to drug therapy in ambulatory patients in the United States costs $76.6 billion annually.31
 

 

Psychiatric disorders including schizophrenia, bipolar disorder, depression, personality disorders, and substance abuse place patients at higher risk for polypharmacy, as do certain demographic, psychosocial, medication, medical, and neurologic factors (Table 4). Other factors that increase the risk for polypharmacy include:

  • institutional factors (recent hospitalization, admission to a surgical ward, nursing home placement, home health care, increased number of pharmacies used, increased number of clinics attended, client-centered psychiatric treatment compared with non-client-centered psychiatric treatment)
  • provider factors (visit to a physician, treatment by general practitioners compared with specialists, increased number of providers, undocumented rationale or diagnosis supporting multiple medication use)
  • having medical insurance.

Steps to avoiding polypharmacy

By identifying polypharmacy’s risk factors, we may decrease its associated morbidity, mortality, and cost. Steps to follow while prescribing—as represented by the mnemonics SAIL33 and TIDE—may help you avoid polypharmacy’s negative consequences.

SAIL. Keep the drug regimen as simple as possible. Aim for once-daily or twice daily dosing. Try to simplify complex drug regimens by discontinuing any drug that does not achieve its defined therapeutic goal. For diseases and syndromes with less clear-cut causes, subtracting drugs from a complicated regimen may be more therapeutic than adding another drug. Try to treat multiple symptoms and syndromes with a single drug that may have multiple beneficial effects, rather than treating each symptom or syndrome with individual drugs.

Understand the potential adverse effects of each drug and potential drug-drug interactions. Whenever practical, choose drugs with broad rather than narrow therapeutic indices.

Each prescribed drug should have a clear indication and a well-defined therapeutic goal. Prescribe using evidence-based medicine as much as is practical.

List the name and dosage of each drug in the patient’s chart, and provide this information to the patient.33 Consider adopting computer data entry and feedback procedures, which have been shown to decrease polypharmacy34 and drug-drug interactions.35

TIDE. In the busy medical practice, writing a prescription signals to the patient that his or her time with the doctor is almost finished. Allow time to address medication issues.

Apply the understanding of individual variability, pharmacokinetics, and pharmacodynamics when prescribing. Review with the patient all prescription and nonprescription drugs and dietary supplements being taken.

Be careful to avoid potentially dangerous drug-drug interactions, especially those associated with serious adverse events such as torsades de pointes.

Educate patients regarding drug and non-drug treatments. Explain potential adverse effects of each drug and potential drug-drug interactions.

Related resources

Disclosure

Drs. Werder and Preskorn have served on the speakers bureau of, as consultants to, or as principal investigators for Abbott Laboratories, AstraZeneca Pharmaceuticals, Biovail Corp., Bristol-Meyers Squibb Co., Merck and Co., Eisai Inc., Eli Lilly and Co., GlaxoSmithKline, Hoffman-LaRoche, Janssen Pharmaceutica, Lundbeck, Novartis Pharmaceuticals Corp., Organon, Pfizer Inc., Solvay, Wyeth Pharmaceuticals, and Yamanouchi Pharmaceuticals Co., Ltd.

“Do no harm” is the first rule of medicine, yet 106,000 Americans die each year from properly prescribed and correctly taken medications.1 In some cases, the cause of death is known and can be attributed to a drug-drug interaction. The likelihood of death or hospitalization is directly proportional to the number of medications a patient is taking, even after controlling for underlying diseases.2

In psychiatry, it is not unusual for us to prescribe more than one psychotropic agent to manage a patient’s symptoms:

  • Patients with affective and psychotic disorders are commonly prescribed combinations of antipsychotics, mood stabilizers, antidepressants (often from more than one class), anxiolytics, antihistamines, and anticholinergics.
  • Patients with posttraumatic stress disorder may take selective serotonin reuptake inhibitors, buspirone, trazodone, antipsychotics, mood stabilizers, benzodiazepines, beta blockers, and opiates.
  • Multiple-drug regimens are used in treating other medical and psychiatric disorders, including chronic pain, fibromyalgia, chronic fatigue syndrome, sleep disorders, and epilepsy.

The greater the number of drugs used, the greater the likelihood that adverse events are emerging and are being treated, sometimes while being mistaken for patient psychopathology. As a prescriber, you are in a unique position to recognize and prevent interactions that can occur when patients are treated with two or more medications. This article defines polypharmacy, describes its consequences, prevalence, and risk factors, and offers an eight-step strategy with two mnemonics to help you avoid adverse events when prescribing multiple-drug regimens.

Box 1

POLYPHARMACY: MANY DRUGS, MANY DEFINITIONS

Poly, from the Greek word polus (many, much) and pharmacy, from the Greek word pharmakon (drug, poison) literally means many drugs or, alternatively, much poison.3 The word polypharmacy first appeared in the medical literature in 1959 in the New England Journal of Medicine4 and in the psychiatric literature in 1969 in an article citing its incidence at a state mental hospital.5

Many definitions have been used to describe and define polypharmacy, both qualitatively and quantitatively. Monotherapy is drug treatment with one drug. Sometimes treatment with two drugs is referred to as co-pharmacy, while treatment with three or more drugs is referred to as polypharmacy.Minor polypharmacy refers to treatment with two to four drugs, while major polypharmacyrefers to treatment with five or more drugs.6

What is polypharmacy?

Many definitions have been used to describe polypharmacy (Box 1).3-6 The most common definition is the use of five or more drugs at the same time in the same patient.7 Although polypharmacy often has a pejorative connotation, using five or more drugs may be therapeutic or contratherapeutic.

Therapeutic polypharmacy occurs, for example, when expert panels or researchers in carefully controlled clinical trials recommend using multiple medications to treat specific diseases. For example, the five-drug combination of isoniazid, rifampin, ethambutol, pyrazinamide, and pyridoxine is therapeutic in initial tuberculosis treatment. More is better in this case because four antibiotics are needed to prevent the development of multiple drug-resistant Mycobacterium tuberculosis, and adding pyridoxine prevents isoniazid-induced neurotoxicity. This example illustrates two prescribing principles:

  • using multiple drugs can help achieve an intended therapeutic goal
  • adding one drug can prevent a known side effect of another drug.

Another example is the therapeutic management of congestive heart failure, in which five drug classes—an angiotensin-converting enzyme (ACE) inhibitor, a diuretic, a digitalis glycoside, a beta blocker, and an aldosterone antagonist—are used in various combinations. All play a role in improving cardiac function and reducing morbidity and mortality.

Using combination drug therapy can also generate cost benefits, such as by adding a drug to delay or inhibit the metabolism of an expensive principal drug. For example, adding diltiazem—a cytochrome P450 (CYP) 3A4 inhibitor—to cyclosporine—which is metabolized by CYP 3A4 enzymes—reduces the dosage of cyclosporine needed to achieve a desired serum level, thereby reducing the cost of this drug. (Some have abandoned this strategy because of cyclosporine’s narrow therapeutic index.)

Contratherapeutic polypharmacy occurs when a patient taking multiple drugs experiences an unexpected or unintended adverse outcome.

Settings for polypharmacy

Polypharmacy occurs in five principal prescribing situations:

  • treatment of symptoms
  • treatment of multiple illnesses
  • treatment of phasic illnesses, such as many affective, anxiety, seizure, and neurodegenerative disorders
  • preventing or treating adverse effects of other drugs
  • attempting to accelerate the onset of action or augment the effects of a preceding drug.

As described above, diseases such as tuberculosis and congestive heart failure, with well-understood causes and pathophysiologies, are often treated with multiple therapeutic drug combinations. However, the causes of many psychiatric disorders and syndromes are less well-understood, which makes prescribing drug combinations more difficult. It may be that treating less well-understood diseases is a risk factor for contratherapeutic polypharmacy.

 

 

Most individuals who are prescribed five or more drugs are taking unique drug combinations.8 These heterogeneous regimens represent “an uncontrolled experiment,” with effects that cannot be predicted from studies in the literature.9Tables 1, 2, and 3 describe how contratherapeutic polypharmacy may occur with combinations of any number of drugs, whether five or more by the classic definition or only two. For example, contratherapeutic polypharmacy may occur when a patient is given the mood-stabilizing drugs valproate and carbamazepine (CBZ) at the same time.10 Here is why this combination may be dangerous:

  • Carbamazepine is oxidized by arene oxidase to CBZ 10,11-epoxide, which is hydrolyzed by epoxide hydrolase to CBZ 10,11-dihydroxide. The metabolite CBZ 10,11-epoxide has both therapeutic and toxic effects.
  • In monotherapy, the ratio of carbamazepine to CBZ 10,11-epoxide is 10:1, with CBZ 10,11-epoxide having a shorter half-life than carbamazepine.
  • However, when carbamazepine and valproate are taken as co-pharmacy, valproate blocks the hydrolysis of CBZ 10,11-epoxide by inhibiting epoxide hydrolase, so that the ratio of carbamazepine to CBZ 10,11-epoxide becomes 2:1. Higher concentrations of the epoxide metabolite contribute to neurotoxicity.

Table 1

POLYPHARMACY WITH TWO OR MORE MEDICATIONS

DescriptionExample
Two or more drugs from the same drug categoryTwo nonsteroidal anti-inflammatory drugs (NSAIDs), two ACE inhibitors, or two phenothiazines
Use of multiple medications across therapeutic classesUse of multiple CNS medications, as in multiple antidepressants, antipsychotics, or anticonvulsants
An inappropriate or unnecessary medication is prescribed to a patient taking other medicationInappropriate prescription due to relative or absolute contraindications Inappropriate prescription due to weak or no indication
Prescription of an exceedingly high dose to a patient taking other medicationThe maximum recommended dose may be functionally exceeded to a serious degree if a drug with a narrow therapeutic index (e.g., amitriptyline) is combined with one that blocks its metabolism (e.g., fluoxetine)
Two or more drugs sharing similar toxicitiesAnticholinergic toxicity due to combining a low-potency phenothiazine antipsychotic and a tertiary amine tricyclic antidepressant

Other examples of potentially dangerous drug combinations include those associated with torsades de pointes, which may occur with certain combinations of antihistamines, antidepressants, antipsychotics, antivirals, antibacterials, antifungals, antiarrhythmics, and promotility agents.

Drug-drug interactions

In a drug-drug interaction, the presence of one drug alters the nature, magnitude, or duration of the effect of a given dose of another drug; the interaction may be either therapeutic or adverse, depending on the desired effect. A drug-drug interaction may be intended or unintended and is determined by pharmacokinetics and pharmacodynamics rather than by therapeutic class.

Most available drug information describes the effects of individual drugs used alone (monopharmacy). Information on how one drug interacts with another (co-pharmacy) is more difficult to come by. A recent literature search using broad criteria for drug-drug interactions uncovered 4,277 indexed articles. Another search, this time using narrow criteria, produced only 316 articles, suggesting that systematic studies regarding drug-drug interactions are few.

Table 2

HOW PHARMACODYNAMICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

MechanismExamples
One drug has a mechanism of action directly opposing the mechanism of action of a co-prescribed drugBromocriptine and prochlorperazine in treating a patient with parkinsonism and nausea
Levidopa/carbidopa and risperidone in treating a patient with parkinsonism and psychosis
Venlafaxine and atenolol in treating a patient with depression and hypertension
One drug has an action that increases the potential for an adverse event of a co-prescribed drugOrthostatic hypotension and syncope when an ACE inhibitor is added to a diuretic
Orthostatic hypotension and syncope when risperidone, because of its action as an alpha-1 adrenergic blocker, is added to a diuretic
Narcosis and respiratory failure when parenteral fentanyl is added to oral meperidine
Neurotoxicity (absence status epilepticus) when valproate is added to clonazepam in children with absence seizures

However, if you understand the pharmacodynamics and pharmacokinetics that rule co-pharmacy, then you can apply this knowledge to more complex drug-drug interactions involving contratherapeutic polypharmacy.

How drug effects are determined. The nature and magnitude of a drug’s effect are determined by its site of action and its binding affinity, concentration, and action at that site.11 This relationship can be represented by the formula:

effect = potency at the site of action × concentration at the site of action

Potency at the site of action is determined by the binding affinity for the drug and the degree to which the receptor is stimulated or blocked, thus activating or inhibiting transmembrane and intracellular messengers (pharmacodynamics). Concentration at the site of action is determined by absorption, metabolism, distribution, and elimination (pharmacokinetics). Thus, the above model can be represented mathematically by:

effect = pharmacodynamics × pharmacokinetics

These factors determine a drug’s usual effect in the usual patient on the usual dosage, which is the goal of most clinical trials. However, all patients are not “usual,” because of inter-individual differences due to genetics, gender, age, environment, social habits such as smoking, intercurrent diseases affecting organ function, and concomitant drug therapy. Thus, when we take these factors into account, the first mathematical equation becomes:

 

 

effect = potency at the site of action × concentration at site of action × inter-individual variance

In other words, the clinical response equals the drug’s potency at the site of action times the drug’s concentration at the site of action times the patient’s underlying biology. Likewise, when we consider variability among patients, the second equation becomes:

effect = pharmacodynamics × pharmacokinetics × inter-individual variance

Table 3

HOW PHARMACOKINETICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

Mechanism of interaction of two or more drugsTwo or more drugs interact where …Examples
One negatively affects the other’s absorption Use of tetracycline with substances containing calcium
One negatively affects the other’s distribution Amiodarone and quinidine, by inhibiting P-glycoprotein, reduce the volume of distribution and/or clearance of digoxin, doubling its serum level
One negatively affects the other’s metabolismOne negatively affects the other’s oxidative metabolism by inducing CYP enzyme activityCarbamazepine induces CYP 2C9 and CYP 3A4 activity, which stimulates warfarin biotransformation, decreases its half-life, and lowers its serum concentration
 One negatively affects the other’s oxidative metabolism by inhibiting CYP enzyme activityKetoconazole inhibits CYP 3A4 activity, which inhibits terfenadine metabolism, resulting in serum terfenadine levels 32 to 100 times normal
 One inhibits hydroxylation of the other’s toxic metabolites, inhibiting their clearanceCombination of carbamazepine and valproate
One negatively affects the other’s elimination Lithium plus hydrochlorothiazide or an NSAID (both impair lithium excretion)

This addition to the equation explains how inter-individual variability can shift the dose-response curve to produce a greater or lesser effect than that which would be expected in the “usual” patient taking the prescribed dosage.

Inter-individual variance. The metabolism of dextromethorphan illustrates the effect of inter-individual variance. After a single dose, about 93% of Caucasians develop relatively lower dextromethorphan:dextrophan ratios, and about 7% develop relatively higher ratios. This difference defines patients who are pharmacogenetically CYP 2D6 extensive metabolizers versus those who are not.

Similarly, drugs sometimes cause biological variance, which predisposes to a drug-drug interaction. For example, the literature is replete with case reports and case series reporting that a substantial CYP 2D6 inhibitor—such as fluoxetine—blocks the metabolism of drugs that are principally metabolized by CYP 2D6. If the drug being metabolized has a narrow therapeutic index—such as amitriptyline—the resultant increase in its serum level can cause serious cardio and neurotoxicity, including arrhythmias, delirium, seizures, coma, and death.12

In such cases, a CYP 2D6 inhibitor converts the phenotype from a CYP 2D6 extensive metabolizer into a CYP 2D6 poor metabolizer. Hence, the clinician must consider how a specific patient may differ from the usual patient when selecting and dosing a drug. The difference may be genetic or acquired, as in this example.

Table 4

RISK FACTORS FOR POLYPHARMACY

Psychiatric disordersMedications being taken
SchizophreniaCardiovascular agents
Bipolar disorderAntipsychotics
DepressionMood stabilizers
Borderline and other personality disordersAntidepressants
Substance abuse (including tobacco habituation)Self-medication with aspirin
Neurologic disordersDemographic variables
Mental retardationAge 65 or older
DementiaEthnicity (Caucasian, African-American)
Chronic pain, facial painFemale gender
Headache (including migraine)Psychosocial variables
InsomniaLower socioeconomic status
EpilepsyInner-city residence
Medical disordersLower level of education
Chronic diseases, multiple diseasesUnemployment
ObesitySelf-medication
DiabetesConcealed drug use
Chronic hypertension 
Coronary artery disease 

The following equation explains how dose is related to drug concentration, which takes into account the drug’s pharmacokinetics:

drug concentration = dosing rate (mg/day) ÷ clearance (ml/min)

In other words, the concentration achieved in a specific patient is determined by the dosage relative to the patient’s ability to clear the drug from the body.

Consequences, prevalence of polypharmacy

Polypharmacy increases patients’ risk for many ill effects, including incidence and severity of adverse events, drug-drug interactions, medication errors, hospitalizations, morbidity, mortality, and direct and indirect costs. At least 12 reports and studies have been published showing the association between polypharmacy and death,2,13-23 and in some of these reports the association is present even after controlling for underlying diseases.

The prevalence of polypharmacy varies by country and population. In Denmark, for example, the prevalence of polypharmacy is approximately 1.2%,6 compared with approximately 7% in the United States.24 Nearly one-half (46%) of all elderly persons admitted to U.S. hospitals may be taking seven or more medications.25 Polypharmacy is especially problematic in patients age 65 and older (Box 2),26-31 in whom the top five preventable threats to health are congestive heart failure, breast cancer, hypertension, pneumonia, and adverse drug events.32 Although older persons make up less than 15% of the population, they take the greatest number and quantity of medications, purchase 40% of all nonprescription medications, and use 33% of all retail prescriptions.30

Box 2

POLYPHARMACY RISKS IN PATIENTS AGE 65 AND OLDER

  • 14% of older patients prescribed psychotropics experience a hip fracture, accounting for 32,000 annual hip fractures in the United States.26
  • 28% of older patients’ hospitalizations are due to adverse events or non-adherence to drug therapy.27
  • 35% of older patients taking three or more prescription medications at hospital discharge are re-hospitalized within 6 months. Problems with medications lead to 6.4% of these re-admissions.28
  • Among older drivers, taking a psychoactive drug multiplies the risk of a motor vehicle accident involving injuries by 1.5 to 5.5 times. The greater the dosage, the greater the risk.29
  • Hospital admissions related to adverse events from medications in older patients cost $20 billion annually (excluding indirect costs).30
  • Morbidity and mortality related to drug therapy in ambulatory patients in the United States costs $76.6 billion annually.31
 

 

Psychiatric disorders including schizophrenia, bipolar disorder, depression, personality disorders, and substance abuse place patients at higher risk for polypharmacy, as do certain demographic, psychosocial, medication, medical, and neurologic factors (Table 4). Other factors that increase the risk for polypharmacy include:

  • institutional factors (recent hospitalization, admission to a surgical ward, nursing home placement, home health care, increased number of pharmacies used, increased number of clinics attended, client-centered psychiatric treatment compared with non-client-centered psychiatric treatment)
  • provider factors (visit to a physician, treatment by general practitioners compared with specialists, increased number of providers, undocumented rationale or diagnosis supporting multiple medication use)
  • having medical insurance.

Steps to avoiding polypharmacy

By identifying polypharmacy’s risk factors, we may decrease its associated morbidity, mortality, and cost. Steps to follow while prescribing—as represented by the mnemonics SAIL33 and TIDE—may help you avoid polypharmacy’s negative consequences.

SAIL. Keep the drug regimen as simple as possible. Aim for once-daily or twice daily dosing. Try to simplify complex drug regimens by discontinuing any drug that does not achieve its defined therapeutic goal. For diseases and syndromes with less clear-cut causes, subtracting drugs from a complicated regimen may be more therapeutic than adding another drug. Try to treat multiple symptoms and syndromes with a single drug that may have multiple beneficial effects, rather than treating each symptom or syndrome with individual drugs.

Understand the potential adverse effects of each drug and potential drug-drug interactions. Whenever practical, choose drugs with broad rather than narrow therapeutic indices.

Each prescribed drug should have a clear indication and a well-defined therapeutic goal. Prescribe using evidence-based medicine as much as is practical.

List the name and dosage of each drug in the patient’s chart, and provide this information to the patient.33 Consider adopting computer data entry and feedback procedures, which have been shown to decrease polypharmacy34 and drug-drug interactions.35

TIDE. In the busy medical practice, writing a prescription signals to the patient that his or her time with the doctor is almost finished. Allow time to address medication issues.

Apply the understanding of individual variability, pharmacokinetics, and pharmacodynamics when prescribing. Review with the patient all prescription and nonprescription drugs and dietary supplements being taken.

Be careful to avoid potentially dangerous drug-drug interactions, especially those associated with serious adverse events such as torsades de pointes.

Educate patients regarding drug and non-drug treatments. Explain potential adverse effects of each drug and potential drug-drug interactions.

Related resources

Disclosure

Drs. Werder and Preskorn have served on the speakers bureau of, as consultants to, or as principal investigators for Abbott Laboratories, AstraZeneca Pharmaceuticals, Biovail Corp., Bristol-Meyers Squibb Co., Merck and Co., Eisai Inc., Eli Lilly and Co., GlaxoSmithKline, Hoffman-LaRoche, Janssen Pharmaceutica, Lundbeck, Novartis Pharmaceuticals Corp., Organon, Pfizer Inc., Solvay, Wyeth Pharmaceuticals, and Yamanouchi Pharmaceuticals Co., Ltd.

References

1. Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 1998;279(15):1200-5.

2. Incalzi RA, Gemma A, Capparella O, et al. Predicting mortality and length of stay of geriatric patients in an acute care general hospital. J Gerontol 1992;47(2):M35-9.

3. Berube MS, Neely DJ, DeVinne PB. American Heritage Dictionary. (2nd College ed). Boston: Houghton Mifflin Co, 1982.

4. Friend DG. Polypharmacy: multiple-ingredient and shotgun prescriptions. N Engl J Med 1959;260(20):1015-8.

5. Sheppard C, Collins L, Fiorentino D, Fracchia J, Merlis S. Polypharmacy in psychiatric treatment. I. Incidence at a state hospital. Curr Ther Res Clin Exp 1969;(12):765-74.

6. Bjerrum L, Rosholm JU, Hallas J, Kragstrup J. Methods for estimating the occurrence of polypharmacy by means of a prescription database. Eur J Clin Pharmacol 1997;53(1):7-11.

7. Werder SF. Polypharmacy: definitions and risk factors (grand rounds). University of Kansas School of Medicine-Wichita, Department of Psychiatry and Behavioral Sciences. Via Christi Regional Medical Center, St. Joseph Campus: Dec 12, 2000.

8. Bjerrum L, Sogaard J, Hallas J, Kragstrup J. Polypharmacy: correlations with sex, age and drug regimen. A prescription database study. Eur J Clin Pharmacol 1998;54(3):197-202.

9. Bjerrum L. Pharmacoepidemiological Studies of Polypharmacy: Methodological issues, population estimates, and influence of practice patterns (PhD thesis). Odense University Faculty of Health Sciences, Department of clinical pharmacology and research unit of general practice. Denmark; 1998. Available at http://www.sdu.dk/health/IPH/genpract/staff/lbjerrum/PHD/PHD.HTM. Accessed Jan. 9, 2003.

10. Wilder BJ. Pharmacokinetics of valproate and carbamazepine. J Clin Psychopharmacol 1992;12(1 suppl):64S-68S.

11. Preskorn SH. The rational basis for the development and use of newer antidepressants. In: Outpatient management of depression: a guide for the practitioner (2nd ed). Caddo, OK: Professional Publications, Inc; 1999;57-103.

12. Vaughan DA. Interaction of fluoxetine with tricyclic antidepressants. Am J Psychiatry 1988;145(11):1478.-

13. Meeker JE, Reynolds PC. Postmortem tissue methamphetamine concentrations following selegiline administration. J Anal Toxicol 1990;14(5):330-1.

14. Sallee FR, DeVane CL, Ferrell RE. Fluoxetine-related death in a child with cytochrome P-450 2D6 genetic deficiency. J Child Adolesc Psychopharmacol 2000;10(1):27-34.

15. Ellis RJ, Mayo MS, Bodensteiner DM. Ciprofloxacin-warfarin coagulopathy: a case series. Am J Hematol 2000;63(1):28-31.

16. Konig SA, Siemes H, Blaker F, et al. Severe hepatotoxicity during valproate therapy: an update and report of eight new fatalities. Epilepsia 1994;35(5):1005-15.

17. Fattinger K, Roos M, Vergeres P, et al. Epidemiology of drug exposure and adverse drug reactions in two Swiss departments of internal medicine. Br J Clin Pharmacol 2000;49(2):158-67.

18. Ebbesen J, Buajordet I, Erikssen J, Svaar H, Brors O, Hilberg T. Drugs as a cause of death. A prospective quality assurance project in a department of medicine (Norwegian). Tidsskr Nor Laegeforen 1995;115(19):2369-72.

19. Alarcon T, Barcena A, Gonzalez-Montalvo JI, Penalosa C, Salgado A. Factors predictive of outcome on admission to an acute geriatric ward. Age Ageing 1999;28(5):429-32.

20. Smith NK, Albazzaz MK. A prospective study of urinary retention and risk of death after proximal femoral fracture. Age Ageing 1996;25(2):150-4.

21. Pulska T, Pahkala K, Laippala P, Kivela SL. Six-year survival of depressed elderly Finns: a community study. Int J Geriatr Psychiatry 1997;12(9):942-50.

22. Waddington JL, Youssef HA, Kinsella A. Mortality in schizophrenia. Antipsychotic polypharmacy and absence of adjunctive anticholinergics over the course of a 10-year prospective study. Br J Psychiatry 1998;173:325-9.

23. Burns R, Nichols LO, Graney MJ, Cloar FT. Impact of continued geriatric outpatient management on health outcomes of older veterans. Arch Intern Med 1995;155(12):1313-8.

24. Kaufman DW, Kelly JP, Rosenberg L, Anderson TE, Mitchell AA. Recent patterns of medication use in the ambulatory adult population of the United States: the Slone survey. JAMA 2002;16;287(3):337-44.

25. Flaherty JH, Perry HM, 3rd, Lynchard GS, Morley JE. Polypharmacy and hospitalization among older home care patients. J Gerontol A Biol Sci Med Sci 2000;55(10):M554-9.

26. Ray WA, Griffin MR, Schaffner W, Baugh DK, Melton LJ, 3rd. Psychotropic drug use and the risk of hip fracture. N Engl J Med 1987;316(7):363-9.

27. Col N, Fanale JE, Kronholm P. The role of medication noncompliance and adverse drug reactions in hospitalizations of the elderly. Arch Intern Med 1990;150(4):841-5.

28. Bero LA, Lipton HL, Bird JA. Characterization of geriatric drug-related hospital readmissions. Med Care 1991;29(10):989-1003.

29. Ray WA, Fought RL, Decker MD. Psychoactive drugs and the risk of injurious motor vehicle crashes in elderly drivers. Am J Epidemiol 1992;136(7):873-83.

30. Prescription drugs and the elderly. Publication AO/HEHS-95-152. Washington, DC: U.S. General Accounting Office, July 1995.

31. Johnson JA, Bootman JL. Drug-related morbidity and mortality. A cost-of-illness model. Arch Intern Med 1995;155(18):1949-56.

32. Fink A, Siu AL, Brook RH, Park RE, Solomon DH. Assuring the quality of health care for older persons. An expert panel’s priorities. JAMA 1987;258(14):1905-8.

33. Lee DR. Polypharmacy: a case report and new protocol for management. J Am Board Fam Pract 1998;11(2):140-4.

34. Hamdy RC, Moore SW, Whalen K, et al. Reducing polypharmacy in extended care. South Med J 1995;88(5):534-8.

35. Haumschild MJ, Ward ES, Bishop JM, Haumschild MS. Pharmacy-based computer system for monitoring and reporting drug interactions. Am J Hosp Pharm 1987;44(2):345-8.

References

1. Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 1998;279(15):1200-5.

2. Incalzi RA, Gemma A, Capparella O, et al. Predicting mortality and length of stay of geriatric patients in an acute care general hospital. J Gerontol 1992;47(2):M35-9.

3. Berube MS, Neely DJ, DeVinne PB. American Heritage Dictionary. (2nd College ed). Boston: Houghton Mifflin Co, 1982.

4. Friend DG. Polypharmacy: multiple-ingredient and shotgun prescriptions. N Engl J Med 1959;260(20):1015-8.

5. Sheppard C, Collins L, Fiorentino D, Fracchia J, Merlis S. Polypharmacy in psychiatric treatment. I. Incidence at a state hospital. Curr Ther Res Clin Exp 1969;(12):765-74.

6. Bjerrum L, Rosholm JU, Hallas J, Kragstrup J. Methods for estimating the occurrence of polypharmacy by means of a prescription database. Eur J Clin Pharmacol 1997;53(1):7-11.

7. Werder SF. Polypharmacy: definitions and risk factors (grand rounds). University of Kansas School of Medicine-Wichita, Department of Psychiatry and Behavioral Sciences. Via Christi Regional Medical Center, St. Joseph Campus: Dec 12, 2000.

8. Bjerrum L, Sogaard J, Hallas J, Kragstrup J. Polypharmacy: correlations with sex, age and drug regimen. A prescription database study. Eur J Clin Pharmacol 1998;54(3):197-202.

9. Bjerrum L. Pharmacoepidemiological Studies of Polypharmacy: Methodological issues, population estimates, and influence of practice patterns (PhD thesis). Odense University Faculty of Health Sciences, Department of clinical pharmacology and research unit of general practice. Denmark; 1998. Available at http://www.sdu.dk/health/IPH/genpract/staff/lbjerrum/PHD/PHD.HTM. Accessed Jan. 9, 2003.

10. Wilder BJ. Pharmacokinetics of valproate and carbamazepine. J Clin Psychopharmacol 1992;12(1 suppl):64S-68S.

11. Preskorn SH. The rational basis for the development and use of newer antidepressants. In: Outpatient management of depression: a guide for the practitioner (2nd ed). Caddo, OK: Professional Publications, Inc; 1999;57-103.

12. Vaughan DA. Interaction of fluoxetine with tricyclic antidepressants. Am J Psychiatry 1988;145(11):1478.-

13. Meeker JE, Reynolds PC. Postmortem tissue methamphetamine concentrations following selegiline administration. J Anal Toxicol 1990;14(5):330-1.

14. Sallee FR, DeVane CL, Ferrell RE. Fluoxetine-related death in a child with cytochrome P-450 2D6 genetic deficiency. J Child Adolesc Psychopharmacol 2000;10(1):27-34.

15. Ellis RJ, Mayo MS, Bodensteiner DM. Ciprofloxacin-warfarin coagulopathy: a case series. Am J Hematol 2000;63(1):28-31.

16. Konig SA, Siemes H, Blaker F, et al. Severe hepatotoxicity during valproate therapy: an update and report of eight new fatalities. Epilepsia 1994;35(5):1005-15.

17. Fattinger K, Roos M, Vergeres P, et al. Epidemiology of drug exposure and adverse drug reactions in two Swiss departments of internal medicine. Br J Clin Pharmacol 2000;49(2):158-67.

18. Ebbesen J, Buajordet I, Erikssen J, Svaar H, Brors O, Hilberg T. Drugs as a cause of death. A prospective quality assurance project in a department of medicine (Norwegian). Tidsskr Nor Laegeforen 1995;115(19):2369-72.

19. Alarcon T, Barcena A, Gonzalez-Montalvo JI, Penalosa C, Salgado A. Factors predictive of outcome on admission to an acute geriatric ward. Age Ageing 1999;28(5):429-32.

20. Smith NK, Albazzaz MK. A prospective study of urinary retention and risk of death after proximal femoral fracture. Age Ageing 1996;25(2):150-4.

21. Pulska T, Pahkala K, Laippala P, Kivela SL. Six-year survival of depressed elderly Finns: a community study. Int J Geriatr Psychiatry 1997;12(9):942-50.

22. Waddington JL, Youssef HA, Kinsella A. Mortality in schizophrenia. Antipsychotic polypharmacy and absence of adjunctive anticholinergics over the course of a 10-year prospective study. Br J Psychiatry 1998;173:325-9.

23. Burns R, Nichols LO, Graney MJ, Cloar FT. Impact of continued geriatric outpatient management on health outcomes of older veterans. Arch Intern Med 1995;155(12):1313-8.

24. Kaufman DW, Kelly JP, Rosenberg L, Anderson TE, Mitchell AA. Recent patterns of medication use in the ambulatory adult population of the United States: the Slone survey. JAMA 2002;16;287(3):337-44.

25. Flaherty JH, Perry HM, 3rd, Lynchard GS, Morley JE. Polypharmacy and hospitalization among older home care patients. J Gerontol A Biol Sci Med Sci 2000;55(10):M554-9.

26. Ray WA, Griffin MR, Schaffner W, Baugh DK, Melton LJ, 3rd. Psychotropic drug use and the risk of hip fracture. N Engl J Med 1987;316(7):363-9.

27. Col N, Fanale JE, Kronholm P. The role of medication noncompliance and adverse drug reactions in hospitalizations of the elderly. Arch Intern Med 1990;150(4):841-5.

28. Bero LA, Lipton HL, Bird JA. Characterization of geriatric drug-related hospital readmissions. Med Care 1991;29(10):989-1003.

29. Ray WA, Fought RL, Decker MD. Psychoactive drugs and the risk of injurious motor vehicle crashes in elderly drivers. Am J Epidemiol 1992;136(7):873-83.

30. Prescription drugs and the elderly. Publication AO/HEHS-95-152. Washington, DC: U.S. General Accounting Office, July 1995.

31. Johnson JA, Bootman JL. Drug-related morbidity and mortality. A cost-of-illness model. Arch Intern Med 1995;155(18):1949-56.

32. Fink A, Siu AL, Brook RH, Park RE, Solomon DH. Assuring the quality of health care for older persons. An expert panel’s priorities. JAMA 1987;258(14):1905-8.

33. Lee DR. Polypharmacy: a case report and new protocol for management. J Am Board Fam Pract 1998;11(2):140-4.

34. Hamdy RC, Moore SW, Whalen K, et al. Reducing polypharmacy in extended care. South Med J 1995;88(5):534-8.

35. Haumschild MJ, Ward ES, Bishop JM, Haumschild MS. Pharmacy-based computer system for monitoring and reporting drug interactions. Am J Hosp Pharm 1987;44(2):345-8.

Issue
Current Psychiatry - 02(02)
Issue
Current Psychiatry - 02(02)
Page Number
24-36
Page Number
24-36
Publications
Publications
Topics
Article Type
Display Headline
Managing polypharmacy: Walking the fine line between help and harm
Display Headline
Managing polypharmacy: Walking the fine line between help and harm
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Managing polypharmacy: Walking the fine line between help and harm

Article Type
Changed
Tue, 12/11/2018 - 15:12
Display Headline
Managing polypharmacy: Walking the fine line between help and harm

“Do no harm” is the first rule of medicine, yet 106,000 Americans die each year from properly prescribed and correctly taken medications.1 In some cases, the cause of death is known and can be attributed to a drug-drug interaction. The likelihood of death or hospitalization is directly proportional to the number of medications a patient is taking, even after controlling for underlying diseases.2

In psychiatry, it is not unusual for us to prescribe more than one psychotropic agent to manage a patient’s symptoms:

  • Patients with affective and psychotic disorders are commonly prescribed combinations of antipsychotics, mood stabilizers, antidepressants (often from more than one class), anxiolytics, antihistamines, and anticholinergics.
  • Patients with posttraumatic stress disorder may take selective serotonin reuptake inhibitors, buspirone, trazodone, antipsychotics, mood stabilizers, benzodiazepines, beta blockers, and opiates.
  • Multiple-drug regimens are used in treating other medical and psychiatric disorders, including chronic pain, fibromyalgia, chronic fatigue syndrome, sleep disorders, and epilepsy.

The greater the number of drugs used, the greater the likelihood that adverse events are emerging and are being treated, sometimes while being mistaken for patient psychopathology. As a prescriber, you are in a unique position to recognize and prevent interactions that can occur when patients are treated with two or more medications. This article defines polypharmacy, describes its consequences, prevalence, and risk factors, and offers an eight-step strategy with two mnemonics to help you avoid adverse events when prescribing multiple-drug regimens.

Box 1

POLYPHARMACY: MANY DRUGS, MANY DEFINITIONS

Poly, from the Greek word polus (many, much) and pharmacy, from the Greek word pharmakon (drug, poison) literally means many drugs or, alternatively, much poison.3 The word polypharmacy first appeared in the medical literature in 1959 in the New England Journal of Medicine4 and in the psychiatric literature in 1969 in an article citing its incidence at a state mental hospital.5

Many definitions have been used to describe and define polypharmacy, both qualitatively and quantitatively. Monotherapy is drug treatment with one drug. Sometimes treatment with two drugs is referred to as co-pharmacy, while treatment with three or more drugs is referred to as polypharmacy.Minor polypharmacy refers to treatment with two to four drugs, while major polypharmacyrefers to treatment with five or more drugs.6

What is polypharmacy?

Many definitions have been used to describe polypharmacy (Box 1).3-6 The most common definition is the use of five or more drugs at the same time in the same patient.7 Although polypharmacy often has a pejorative connotation, using five or more drugs may be therapeutic or contratherapeutic.

Therapeutic polypharmacy occurs, for example, when expert panels or researchers in carefully controlled clinical trials recommend using multiple medications to treat specific diseases. For example, the five-drug combination of isoniazid, rifampin, ethambutol, pyrazinamide, and pyridoxine is therapeutic in initial tuberculosis treatment. More is better in this case because four antibiotics are needed to prevent the development of multiple drug-resistant Mycobacterium tuberculosis, and adding pyridoxine prevents isoniazid-induced neurotoxicity. This example illustrates two prescribing principles:

  • using multiple drugs can help achieve an intended therapeutic goal
  • adding one drug can prevent a known side effect of another drug.

Another example is the therapeutic management of congestive heart failure, in which five drug classes—an angiotensin-converting enzyme (ACE) inhibitor, a diuretic, a digitalis glycoside, a beta blocker, and an aldosterone antagonist—are used in various combinations. All play a role in improving cardiac function and reducing morbidity and mortality.

Using combination drug therapy can also generate cost benefits, such as by adding a drug to delay or inhibit the metabolism of an expensive principal drug. For example, adding diltiazem—a cytochrome P450 (CYP) 3A4 inhibitor—to cyclosporine—which is metabolized by CYP 3A4 enzymes—reduces the dosage of cyclosporine needed to achieve a desired serum level, thereby reducing the cost of this drug. (Some have abandoned this strategy because of cyclosporine’s narrow therapeutic index.)

Contratherapeutic polypharmacy occurs when a patient taking multiple drugs experiences an unexpected or unintended adverse outcome.

Settings for polypharmacy

Polypharmacy occurs in five principal prescribing situations:

  • treatment of symptoms
  • treatment of multiple illnesses
  • treatment of phasic illnesses, such as many affective, anxiety, seizure, and neurodegenerative disorders
  • preventing or treating adverse effects of other drugs
  • attempting to accelerate the onset of action or augment the effects of a preceding drug.

As described above, diseases such as tuberculosis and congestive heart failure, with well-understood causes and pathophysiologies, are often treated with multiple therapeutic drug combinations. However, the causes of many psychiatric disorders and syndromes are less well-understood, which makes prescribing drug combinations more difficult. It may be that treating less well-understood diseases is a risk factor for contratherapeutic polypharmacy.

 

 

Most individuals who are prescribed five or more drugs are taking unique drug combinations.8 These heterogeneous regimens represent “an uncontrolled experiment,” with effects that cannot be predicted from studies in the literature.9Tables 1, 2, and 3 describe how contratherapeutic polypharmacy may occur with combinations of any number of drugs, whether five or more by the classic definition or only two. For example, contratherapeutic polypharmacy may occur when a patient is given the mood-stabilizing drugs valproate and carbamazepine (CBZ) at the same time.10 Here is why this combination may be dangerous:

  • Carbamazepine is oxidized by arene oxidase to CBZ 10,11-epoxide, which is hydrolyzed by epoxide hydrolase to CBZ 10,11-dihydroxide. The metabolite CBZ 10,11-epoxide has both therapeutic and toxic effects.
  • In monotherapy, the ratio of carbamazepine to CBZ 10,11-epoxide is 10:1, with CBZ 10,11-epoxide having a shorter half-life than carbamazepine.
  • However, when carbamazepine and valproate are taken as co-pharmacy, valproate blocks the hydrolysis of CBZ 10,11-epoxide by inhibiting epoxide hydrolase, so that the ratio of carbamazepine to CBZ 10,11-epoxide becomes 2:1. Higher concentrations of the epoxide metabolite contribute to neurotoxicity.

Table 1

POLYPHARMACY WITH TWO OR MORE MEDICATIONS

DescriptionExample
Two or more drugs from the same drug categoryTwo nonsteroidal anti-inflammatory drugs (NSAIDs), two ACE inhibitors, or two phenothiazines
Use of multiple medications across therapeutic classesUse of multiple CNS medications, as in multiple antidepressants, antipsychotics, or anticonvulsants
An inappropriate or unnecessary medication is prescribed to a patient taking other medicationInappropriate prescription due to relative or absolute contraindications Inappropriate prescription due to weak or no indication
Prescription of an exceedingly high dose to a patient taking other medicationThe maximum recommended dose may be functionally exceeded to a serious degree if a drug with a narrow therapeutic index (e.g., amitriptyline) is combined with one that blocks its metabolism (e.g., fluoxetine)
Two or more drugs sharing similar toxicitiesAnticholinergic toxicity due to combining a low-potency phenothiazine antipsychotic and a tertiary amine tricyclic antidepressant

Other examples of potentially dangerous drug combinations include those associated with torsades de pointes, which may occur with certain combinations of antihistamines, antidepressants, antipsychotics, antivirals, antibacterials, antifungals, antiarrhythmics, and promotility agents.

Drug-drug interactions

In a drug-drug interaction, the presence of one drug alters the nature, magnitude, or duration of the effect of a given dose of another drug; the interaction may be either therapeutic or adverse, depending on the desired effect. A drug-drug interaction may be intended or unintended and is determined by pharmacokinetics and pharmacodynamics rather than by therapeutic class.

Most available drug information describes the effects of individual drugs used alone (monopharmacy). Information on how one drug interacts with another (co-pharmacy) is more difficult to come by. A recent literature search using broad criteria for drug-drug interactions uncovered 4,277 indexed articles. Another search, this time using narrow criteria, produced only 316 articles, suggesting that systematic studies regarding drug-drug interactions are few.

Table 2

HOW PHARMACODYNAMICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

MechanismExamples
One drug has a mechanism of action directly opposing the mechanism of action of a co-prescribed drugBromocriptine and prochlorperazine in treating a patient with parkinsonism and nausea
Levidopa/carbidopa and risperidone in treating a patient with parkinsonism and psychosis
Venlafaxine and atenolol in treating a patient with depression and hypertension
One drug has an action that increases the potential for an adverse event of a co-prescribed drugOrthostatic hypotension and syncope when an ACE inhibitor is added to a diuretic
Orthostatic hypotension and syncope when risperidone, because of its action as an alpha-1 adrenergic blocker, is added to a diuretic
Narcosis and respiratory failure when parenteral fentanyl is added to oral meperidine
Neurotoxicity (absence status epilepticus) when valproate is added to clonazepam in children with absence seizures

However, if you understand the pharmacodynamics and pharmacokinetics that rule co-pharmacy, then you can apply this knowledge to more complex drug-drug interactions involving contratherapeutic polypharmacy.

How drug effects are determined. The nature and magnitude of a drug’s effect are determined by its site of action and its binding affinity, concentration, and action at that site.11 This relationship can be represented by the formula:

effect = potency at the site of action × concentration at the site of action

Potency at the site of action is determined by the binding affinity for the drug and the degree to which the receptor is stimulated or blocked, thus activating or inhibiting transmembrane and intracellular messengers (pharmacodynamics). Concentration at the site of action is determined by absorption, metabolism, distribution, and elimination (pharmacokinetics). Thus, the above model can be represented mathematically by:

effect = pharmacodynamics × pharmacokinetics

These factors determine a drug’s usual effect in the usual patient on the usual dosage, which is the goal of most clinical trials. However, all patients are not “usual,” because of inter-individual differences due to genetics, gender, age, environment, social habits such as smoking, intercurrent diseases affecting organ function, and concomitant drug therapy. Thus, when we take these factors into account, the first mathematical equation becomes:

 

 

effect = potency at the site of action × concentration at site of action × inter-individual variance

In other words, the clinical response equals the drug’s potency at the site of action times the drug’s concentration at the site of action times the patient’s underlying biology. Likewise, when we consider variability among patients, the second equation becomes:

effect = pharmacodynamics × pharmacokinetics × inter-individual variance

Table 3

HOW PHARMACOKINETICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

Mechanism of interaction of two or more drugsTwo or more drugs interact where …Examples
One negatively affects the other’s absorption Use of tetracycline with substances containing calcium
One negatively affects the other’s distribution Amiodarone and quinidine, by inhibiting P-glycoprotein, reduce the volume of distribution and/or clearance of digoxin, doubling its serum level
One negatively affects the other’s metabolismOne negatively affects the other’s oxidative metabolism by inducing CYP enzyme activityCarbamazepine induces CYP 2C9 and CYP 3A4 activity, which stimulates warfarin biotransformation, decreases its half-life, and lowers its serum concentration
 One negatively affects the other’s oxidative metabolism by inhibiting CYP enzyme activityKetoconazole inhibits CYP 3A4 activity, which inhibits terfenadine metabolism, resulting in serum terfenadine levels 32 to 100 times normal
 One inhibits hydroxylation of the other’s toxic metabolites, inhibiting their clearanceCombination of carbamazepine and valproate
One negatively affects the other’s elimination Lithium plus hydrochlorothiazide or an NSAID (both impair lithium excretion)

This addition to the equation explains how inter-individual variability can shift the dose-response curve to produce a greater or lesser effect than that which would be expected in the “usual” patient taking the prescribed dosage.

Inter-individual variance. The metabolism of dextromethorphan illustrates the effect of inter-individual variance. After a single dose, about 93% of Caucasians develop relatively lower dextromethorphan:dextrophan ratios, and about 7% develop relatively higher ratios. This difference defines patients who are pharmacogenetically CYP 2D6 extensive metabolizers versus those who are not.

Similarly, drugs sometimes cause biological variance, which predisposes to a drug-drug interaction. For example, the literature is replete with case reports and case series reporting that a substantial CYP 2D6 inhibitor—such as fluoxetine—blocks the metabolism of drugs that are principally metabolized by CYP 2D6. If the drug being metabolized has a narrow therapeutic index—such as amitriptyline—the resultant increase in its serum level can cause serious cardio and neurotoxicity, including arrhythmias, delirium, seizures, coma, and death.12

In such cases, a CYP 2D6 inhibitor converts the phenotype from a CYP 2D6 extensive metabolizer into a CYP 2D6 poor metabolizer. Hence, the clinician must consider how a specific patient may differ from the usual patient when selecting and dosing a drug. The difference may be genetic or acquired, as in this example.

Table 4

RISK FACTORS FOR POLYPHARMACY

Psychiatric disordersMedications being taken
SchizophreniaCardiovascular agents
Bipolar disorderAntipsychotics
DepressionMood stabilizers
Borderline and other personality disordersAntidepressants
Substance abuse (including tobacco habituation)Self-medication with aspirin
Neurologic disordersDemographic variables
Mental retardationAge 65 or older
DementiaEthnicity (Caucasian, African-American)
Chronic pain, facial painFemale gender
Headache (including migraine)Psychosocial variables
InsomniaLower socioeconomic status
EpilepsyInner-city residence
Medical disordersLower level of education
Chronic diseases, multiple diseasesUnemployment
ObesitySelf-medication
DiabetesConcealed drug use
Chronic hypertension 
Coronary artery disease 

The following equation explains how dose is related to drug concentration, which takes into account the drug’s pharmacokinetics:

drug concentration = dosing rate (mg/day) ÷ clearance (ml/min)

In other words, the concentration achieved in a specific patient is determined by the dosage relative to the patient’s ability to clear the drug from the body.

Consequences, prevalence of polypharmacy

Polypharmacy increases patients’ risk for many ill effects, including incidence and severity of adverse events, drug-drug interactions, medication errors, hospitalizations, morbidity, mortality, and direct and indirect costs. At least 12 reports and studies have been published showing the association between polypharmacy and death,2,13-23 and in some of these reports the association is present even after controlling for underlying diseases.

The prevalence of polypharmacy varies by country and population. In Denmark, for example, the prevalence of polypharmacy is approximately 1.2%,6 compared with approximately 7% in the United States.24 Nearly one-half (46%) of all elderly persons admitted to U.S. hospitals may be taking seven or more medications.25 Polypharmacy is especially problematic in patients age 65 and older (Box 2),26-31 in whom the top five preventable threats to health are congestive heart failure, breast cancer, hypertension, pneumonia, and adverse drug events.32 Although older persons make up less than 15% of the population, they take the greatest number and quantity of medications, purchase 40% of all nonprescription medications, and use 33% of all retail prescriptions.30

Box 2

POLYPHARMACY RISKS IN PATIENTS AGE 65 AND OLDER

  • 14% of older patients prescribed psychotropics experience a hip fracture, accounting for 32,000 annual hip fractures in the United States.26
  • 28% of older patients’ hospitalizations are due to adverse events or non-adherence to drug therapy.27
  • 35% of older patients taking three or more prescription medications at hospital discharge are re-hospitalized within 6 months. Problems with medications lead to 6.4% of these re-admissions.28
  • Among older drivers, taking a psychoactive drug multiplies the risk of a motor vehicle accident involving injuries by 1.5 to 5.5 times. The greater the dosage, the greater the risk.29
  • Hospital admissions related to adverse events from medications in older patients cost $20 billion annually (excluding indirect costs).30
  • Morbidity and mortality related to drug therapy in ambulatory patients in the United States costs $76.6 billion annually.31
 

 

Psychiatric disorders including schizophrenia, bipolar disorder, depression, personality disorders, and substance abuse place patients at higher risk for polypharmacy, as do certain demographic, psychosocial, medication, medical, and neurologic factors (Table 4). Other factors that increase the risk for polypharmacy include:

  • institutional factors (recent hospitalization, admission to a surgical ward, nursing home placement, home health care, increased number of pharmacies used, increased number of clinics attended, client-centered psychiatric treatment compared with non-client-centered psychiatric treatment)
  • provider factors (visit to a physician, treatment by general practitioners compared with specialists, increased number of providers, undocumented rationale or diagnosis supporting multiple medication use)
  • having medical insurance.

Steps to avoiding polypharmacy

By identifying polypharmacy’s risk factors, we may decrease its associated morbidity, mortality, and cost. Steps to follow while prescribing—as represented by the mnemonics SAIL33 and TIDE—may help you avoid polypharmacy’s negative consequences.

SAIL. Keep the drug regimen as simple as possible. Aim for once-daily or twice daily dosing. Try to simplify complex drug regimens by discontinuing any drug that does not achieve its defined therapeutic goal. For diseases and syndromes with less clear-cut causes, subtracting drugs from a complicated regimen may be more therapeutic than adding another drug. Try to treat multiple symptoms and syndromes with a single drug that may have multiple beneficial effects, rather than treating each symptom or syndrome with individual drugs.

Understand the potential adverse effects of each drug and potential drug-drug interactions. Whenever practical, choose drugs with broad rather than narrow therapeutic indices.

Each prescribed drug should have a clear indication and a well-defined therapeutic goal. Prescribe using evidence-based medicine as much as is practical.

List the name and dosage of each drug in the patient’s chart, and provide this information to the patient.33 Consider adopting computer data entry and feedback procedures, which have been shown to decrease polypharmacy34 and drug-drug interactions.35

TIDE. In the busy medical practice, writing a prescription signals to the patient that his or her time with the doctor is almost finished. Allow time to address medication issues.

Apply the understanding of individual variability, pharmacokinetics, and pharmacodynamics when prescribing. Review with the patient all prescription and nonprescription drugs and dietary supplements being taken.

Be careful to avoid potentially dangerous drug-drug interactions, especially those associated with serious adverse events such as torsades de pointes.

Educate patients regarding drug and non-drug treatments. Explain potential adverse effects of each drug and potential drug-drug interactions.

Related resources

Disclosure

Drs. Werder and Preskorn have served on the speakers bureau of, as consultants to, or as principal investigators for Abbott Laboratories, AstraZeneca Pharmaceuticals, Biovail Corp., Bristol-Meyers Squibb Co., Merck and Co., Eisai Inc., Eli Lilly and Co., GlaxoSmithKline, Hoffman-LaRoche, Janssen Pharmaceutica, Lundbeck, Novartis Pharmaceuticals Corp., Organon, Pfizer Inc., Solvay, Wyeth Pharmaceuticals, and Yamanouchi Pharmaceuticals Co., Ltd.

References

1. Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 1998;279(15):1200-5.

2. Incalzi RA, Gemma A, Capparella O, et al. Predicting mortality and length of stay of geriatric patients in an acute care general hospital. J Gerontol 1992;47(2):M35-9.

3. Berube MS, Neely DJ, DeVinne PB. American Heritage Dictionary. (2nd College ed). Boston: Houghton Mifflin Co, 1982.

4. Friend DG. Polypharmacy: multiple-ingredient and shotgun prescriptions. N Engl J Med 1959;260(20):1015-8.

5. Sheppard C, Collins L, Fiorentino D, Fracchia J, Merlis S. Polypharmacy in psychiatric treatment. I. Incidence at a state hospital. Curr Ther Res Clin Exp 1969;(12):765-74.

6. Bjerrum L, Rosholm JU, Hallas J, Kragstrup J. Methods for estimating the occurrence of polypharmacy by means of a prescription database. Eur J Clin Pharmacol 1997;53(1):7-11.

7. Werder SF. Polypharmacy: definitions and risk factors (grand rounds). University of Kansas School of Medicine-Wichita, Department of Psychiatry and Behavioral Sciences. Via Christi Regional Medical Center, St. Joseph Campus: Dec 12, 2000.

8. Bjerrum L, Sogaard J, Hallas J, Kragstrup J. Polypharmacy: correlations with sex, age and drug regimen. A prescription database study. Eur J Clin Pharmacol 1998;54(3):197-202.

9. Bjerrum L. Pharmacoepidemiological Studies of Polypharmacy: Methodological issues, population estimates, and influence of practice patterns (PhD thesis). Odense University Faculty of Health Sciences, Department of clinical pharmacology and research unit of general practice. Denmark; 1998. Available at http://www.sdu.dk/health/IPH/genpract/staff/lbjerrum/PHD/PHD.HTM. Accessed Jan. 9, 2003.

10. Wilder BJ. Pharmacokinetics of valproate and carbamazepine. J Clin Psychopharmacol 1992;12(1 suppl):64S-68S.

11. Preskorn SH. The rational basis for the development and use of newer antidepressants. In: Outpatient management of depression: a guide for the practitioner (2nd ed). Caddo, OK: Professional Publications, Inc; 1999;57-103.

12. Vaughan DA. Interaction of fluoxetine with tricyclic antidepressants. Am J Psychiatry 1988;145(11):1478.-

13. Meeker JE, Reynolds PC. Postmortem tissue methamphetamine concentrations following selegiline administration. J Anal Toxicol 1990;14(5):330-1.

14. Sallee FR, DeVane CL, Ferrell RE. Fluoxetine-related death in a child with cytochrome P-450 2D6 genetic deficiency. J Child Adolesc Psychopharmacol 2000;10(1):27-34.

15. Ellis RJ, Mayo MS, Bodensteiner DM. Ciprofloxacin-warfarin coagulopathy: a case series. Am J Hematol 2000;63(1):28-31.

16. Konig SA, Siemes H, Blaker F, et al. Severe hepatotoxicity during valproate therapy: an update and report of eight new fatalities. Epilepsia 1994;35(5):1005-15.

17. Fattinger K, Roos M, Vergeres P, et al. Epidemiology of drug exposure and adverse drug reactions in two Swiss departments of internal medicine. Br J Clin Pharmacol 2000;49(2):158-67.

18. Ebbesen J, Buajordet I, Erikssen J, Svaar H, Brors O, Hilberg T. Drugs as a cause of death. A prospective quality assurance project in a department of medicine (Norwegian). Tidsskr Nor Laegeforen 1995;115(19):2369-72.

19. Alarcon T, Barcena A, Gonzalez-Montalvo JI, Penalosa C, Salgado A. Factors predictive of outcome on admission to an acute geriatric ward. Age Ageing 1999;28(5):429-32.

20. Smith NK, Albazzaz MK. A prospective study of urinary retention and risk of death after proximal femoral fracture. Age Ageing 1996;25(2):150-4.

21. Pulska T, Pahkala K, Laippala P, Kivela SL. Six-year survival of depressed elderly Finns: a community study. Int J Geriatr Psychiatry 1997;12(9):942-50.

22. Waddington JL, Youssef HA, Kinsella A. Mortality in schizophrenia. Antipsychotic polypharmacy and absence of adjunctive anticholinergics over the course of a 10-year prospective study. Br J Psychiatry 1998;173:325-9.

23. Burns R, Nichols LO, Graney MJ, Cloar FT. Impact of continued geriatric outpatient management on health outcomes of older veterans. Arch Intern Med 1995;155(12):1313-8.

24. Kaufman DW, Kelly JP, Rosenberg L, Anderson TE, Mitchell AA. Recent patterns of medication use in the ambulatory adult population of the United States: the Slone survey. JAMA 2002;16;287(3):337-44.

25. Flaherty JH, Perry HM, 3rd, Lynchard GS, Morley JE. Polypharmacy and hospitalization among older home care patients. J Gerontol A Biol Sci Med Sci 2000;55(10):M554-9.

26. Ray WA, Griffin MR, Schaffner W, Baugh DK, Melton LJ, 3rd. Psychotropic drug use and the risk of hip fracture. N Engl J Med 1987;316(7):363-9.

27. Col N, Fanale JE, Kronholm P. The role of medication noncompliance and adverse drug reactions in hospitalizations of the elderly. Arch Intern Med 1990;150(4):841-5.

28. Bero LA, Lipton HL, Bird JA. Characterization of geriatric drug-related hospital readmissions. Med Care 1991;29(10):989-1003.

29. Ray WA, Fought RL, Decker MD. Psychoactive drugs and the risk of injurious motor vehicle crashes in elderly drivers. Am J Epidemiol 1992;136(7):873-83.

30. Prescription drugs and the elderly. Publication AO/HEHS-95-152. Washington, DC: U.S. General Accounting Office, July 1995.

31. Johnson JA, Bootman JL. Drug-related morbidity and mortality. A cost-of-illness model. Arch Intern Med 1995;155(18):1949-56.

32. Fink A, Siu AL, Brook RH, Park RE, Solomon DH. Assuring the quality of health care for older persons. An expert panel’s priorities. JAMA 1987;258(14):1905-8.

33. Lee DR. Polypharmacy: a case report and new protocol for management. J Am Board Fam Pract 1998;11(2):140-4.

34. Hamdy RC, Moore SW, Whalen K, et al. Reducing polypharmacy in extended care. South Med J 1995;88(5):534-8.

35. Haumschild MJ, Ward ES, Bishop JM, Haumschild MS. Pharmacy-based computer system for monitoring and reporting drug interactions. Am J Hosp Pharm 1987;44(2):345-8.

Article PDF
Author and Disclosure Information

Steven F. Werder, DO
Assistant professor Departments of psychiatry and internal medicine University of Kansas School of Medicine Wichita

Sheldon H. Preskorn, MD
Professor and chairman Department of psychiatry and behavioral sciences

University of Kansas School of Medicine Wichita

Issue
Current Psychiatry - 02(02)
Publications
Topics
Page Number
24-36
Sections
Author and Disclosure Information

Steven F. Werder, DO
Assistant professor Departments of psychiatry and internal medicine University of Kansas School of Medicine Wichita

Sheldon H. Preskorn, MD
Professor and chairman Department of psychiatry and behavioral sciences

University of Kansas School of Medicine Wichita

Author and Disclosure Information

Steven F. Werder, DO
Assistant professor Departments of psychiatry and internal medicine University of Kansas School of Medicine Wichita

Sheldon H. Preskorn, MD
Professor and chairman Department of psychiatry and behavioral sciences

University of Kansas School of Medicine Wichita

Article PDF
Article PDF

“Do no harm” is the first rule of medicine, yet 106,000 Americans die each year from properly prescribed and correctly taken medications.1 In some cases, the cause of death is known and can be attributed to a drug-drug interaction. The likelihood of death or hospitalization is directly proportional to the number of medications a patient is taking, even after controlling for underlying diseases.2

In psychiatry, it is not unusual for us to prescribe more than one psychotropic agent to manage a patient’s symptoms:

  • Patients with affective and psychotic disorders are commonly prescribed combinations of antipsychotics, mood stabilizers, antidepressants (often from more than one class), anxiolytics, antihistamines, and anticholinergics.
  • Patients with posttraumatic stress disorder may take selective serotonin reuptake inhibitors, buspirone, trazodone, antipsychotics, mood stabilizers, benzodiazepines, beta blockers, and opiates.
  • Multiple-drug regimens are used in treating other medical and psychiatric disorders, including chronic pain, fibromyalgia, chronic fatigue syndrome, sleep disorders, and epilepsy.

The greater the number of drugs used, the greater the likelihood that adverse events are emerging and are being treated, sometimes while being mistaken for patient psychopathology. As a prescriber, you are in a unique position to recognize and prevent interactions that can occur when patients are treated with two or more medications. This article defines polypharmacy, describes its consequences, prevalence, and risk factors, and offers an eight-step strategy with two mnemonics to help you avoid adverse events when prescribing multiple-drug regimens.

Box 1

POLYPHARMACY: MANY DRUGS, MANY DEFINITIONS

Poly, from the Greek word polus (many, much) and pharmacy, from the Greek word pharmakon (drug, poison) literally means many drugs or, alternatively, much poison.3 The word polypharmacy first appeared in the medical literature in 1959 in the New England Journal of Medicine4 and in the psychiatric literature in 1969 in an article citing its incidence at a state mental hospital.5

Many definitions have been used to describe and define polypharmacy, both qualitatively and quantitatively. Monotherapy is drug treatment with one drug. Sometimes treatment with two drugs is referred to as co-pharmacy, while treatment with three or more drugs is referred to as polypharmacy.Minor polypharmacy refers to treatment with two to four drugs, while major polypharmacyrefers to treatment with five or more drugs.6

What is polypharmacy?

Many definitions have been used to describe polypharmacy (Box 1).3-6 The most common definition is the use of five or more drugs at the same time in the same patient.7 Although polypharmacy often has a pejorative connotation, using five or more drugs may be therapeutic or contratherapeutic.

Therapeutic polypharmacy occurs, for example, when expert panels or researchers in carefully controlled clinical trials recommend using multiple medications to treat specific diseases. For example, the five-drug combination of isoniazid, rifampin, ethambutol, pyrazinamide, and pyridoxine is therapeutic in initial tuberculosis treatment. More is better in this case because four antibiotics are needed to prevent the development of multiple drug-resistant Mycobacterium tuberculosis, and adding pyridoxine prevents isoniazid-induced neurotoxicity. This example illustrates two prescribing principles:

  • using multiple drugs can help achieve an intended therapeutic goal
  • adding one drug can prevent a known side effect of another drug.

Another example is the therapeutic management of congestive heart failure, in which five drug classes—an angiotensin-converting enzyme (ACE) inhibitor, a diuretic, a digitalis glycoside, a beta blocker, and an aldosterone antagonist—are used in various combinations. All play a role in improving cardiac function and reducing morbidity and mortality.

Using combination drug therapy can also generate cost benefits, such as by adding a drug to delay or inhibit the metabolism of an expensive principal drug. For example, adding diltiazem—a cytochrome P450 (CYP) 3A4 inhibitor—to cyclosporine—which is metabolized by CYP 3A4 enzymes—reduces the dosage of cyclosporine needed to achieve a desired serum level, thereby reducing the cost of this drug. (Some have abandoned this strategy because of cyclosporine’s narrow therapeutic index.)

Contratherapeutic polypharmacy occurs when a patient taking multiple drugs experiences an unexpected or unintended adverse outcome.

Settings for polypharmacy

Polypharmacy occurs in five principal prescribing situations:

  • treatment of symptoms
  • treatment of multiple illnesses
  • treatment of phasic illnesses, such as many affective, anxiety, seizure, and neurodegenerative disorders
  • preventing or treating adverse effects of other drugs
  • attempting to accelerate the onset of action or augment the effects of a preceding drug.

As described above, diseases such as tuberculosis and congestive heart failure, with well-understood causes and pathophysiologies, are often treated with multiple therapeutic drug combinations. However, the causes of many psychiatric disorders and syndromes are less well-understood, which makes prescribing drug combinations more difficult. It may be that treating less well-understood diseases is a risk factor for contratherapeutic polypharmacy.

 

 

Most individuals who are prescribed five or more drugs are taking unique drug combinations.8 These heterogeneous regimens represent “an uncontrolled experiment,” with effects that cannot be predicted from studies in the literature.9Tables 1, 2, and 3 describe how contratherapeutic polypharmacy may occur with combinations of any number of drugs, whether five or more by the classic definition or only two. For example, contratherapeutic polypharmacy may occur when a patient is given the mood-stabilizing drugs valproate and carbamazepine (CBZ) at the same time.10 Here is why this combination may be dangerous:

  • Carbamazepine is oxidized by arene oxidase to CBZ 10,11-epoxide, which is hydrolyzed by epoxide hydrolase to CBZ 10,11-dihydroxide. The metabolite CBZ 10,11-epoxide has both therapeutic and toxic effects.
  • In monotherapy, the ratio of carbamazepine to CBZ 10,11-epoxide is 10:1, with CBZ 10,11-epoxide having a shorter half-life than carbamazepine.
  • However, when carbamazepine and valproate are taken as co-pharmacy, valproate blocks the hydrolysis of CBZ 10,11-epoxide by inhibiting epoxide hydrolase, so that the ratio of carbamazepine to CBZ 10,11-epoxide becomes 2:1. Higher concentrations of the epoxide metabolite contribute to neurotoxicity.

Table 1

POLYPHARMACY WITH TWO OR MORE MEDICATIONS

DescriptionExample
Two or more drugs from the same drug categoryTwo nonsteroidal anti-inflammatory drugs (NSAIDs), two ACE inhibitors, or two phenothiazines
Use of multiple medications across therapeutic classesUse of multiple CNS medications, as in multiple antidepressants, antipsychotics, or anticonvulsants
An inappropriate or unnecessary medication is prescribed to a patient taking other medicationInappropriate prescription due to relative or absolute contraindications Inappropriate prescription due to weak or no indication
Prescription of an exceedingly high dose to a patient taking other medicationThe maximum recommended dose may be functionally exceeded to a serious degree if a drug with a narrow therapeutic index (e.g., amitriptyline) is combined with one that blocks its metabolism (e.g., fluoxetine)
Two or more drugs sharing similar toxicitiesAnticholinergic toxicity due to combining a low-potency phenothiazine antipsychotic and a tertiary amine tricyclic antidepressant

Other examples of potentially dangerous drug combinations include those associated with torsades de pointes, which may occur with certain combinations of antihistamines, antidepressants, antipsychotics, antivirals, antibacterials, antifungals, antiarrhythmics, and promotility agents.

Drug-drug interactions

In a drug-drug interaction, the presence of one drug alters the nature, magnitude, or duration of the effect of a given dose of another drug; the interaction may be either therapeutic or adverse, depending on the desired effect. A drug-drug interaction may be intended or unintended and is determined by pharmacokinetics and pharmacodynamics rather than by therapeutic class.

Most available drug information describes the effects of individual drugs used alone (monopharmacy). Information on how one drug interacts with another (co-pharmacy) is more difficult to come by. A recent literature search using broad criteria for drug-drug interactions uncovered 4,277 indexed articles. Another search, this time using narrow criteria, produced only 316 articles, suggesting that systematic studies regarding drug-drug interactions are few.

Table 2

HOW PHARMACODYNAMICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

MechanismExamples
One drug has a mechanism of action directly opposing the mechanism of action of a co-prescribed drugBromocriptine and prochlorperazine in treating a patient with parkinsonism and nausea
Levidopa/carbidopa and risperidone in treating a patient with parkinsonism and psychosis
Venlafaxine and atenolol in treating a patient with depression and hypertension
One drug has an action that increases the potential for an adverse event of a co-prescribed drugOrthostatic hypotension and syncope when an ACE inhibitor is added to a diuretic
Orthostatic hypotension and syncope when risperidone, because of its action as an alpha-1 adrenergic blocker, is added to a diuretic
Narcosis and respiratory failure when parenteral fentanyl is added to oral meperidine
Neurotoxicity (absence status epilepticus) when valproate is added to clonazepam in children with absence seizures

However, if you understand the pharmacodynamics and pharmacokinetics that rule co-pharmacy, then you can apply this knowledge to more complex drug-drug interactions involving contratherapeutic polypharmacy.

How drug effects are determined. The nature and magnitude of a drug’s effect are determined by its site of action and its binding affinity, concentration, and action at that site.11 This relationship can be represented by the formula:

effect = potency at the site of action × concentration at the site of action

Potency at the site of action is determined by the binding affinity for the drug and the degree to which the receptor is stimulated or blocked, thus activating or inhibiting transmembrane and intracellular messengers (pharmacodynamics). Concentration at the site of action is determined by absorption, metabolism, distribution, and elimination (pharmacokinetics). Thus, the above model can be represented mathematically by:

effect = pharmacodynamics × pharmacokinetics

These factors determine a drug’s usual effect in the usual patient on the usual dosage, which is the goal of most clinical trials. However, all patients are not “usual,” because of inter-individual differences due to genetics, gender, age, environment, social habits such as smoking, intercurrent diseases affecting organ function, and concomitant drug therapy. Thus, when we take these factors into account, the first mathematical equation becomes:

 

 

effect = potency at the site of action × concentration at site of action × inter-individual variance

In other words, the clinical response equals the drug’s potency at the site of action times the drug’s concentration at the site of action times the patient’s underlying biology. Likewise, when we consider variability among patients, the second equation becomes:

effect = pharmacodynamics × pharmacokinetics × inter-individual variance

Table 3

HOW PHARMACOKINETICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

Mechanism of interaction of two or more drugsTwo or more drugs interact where …Examples
One negatively affects the other’s absorption Use of tetracycline with substances containing calcium
One negatively affects the other’s distribution Amiodarone and quinidine, by inhibiting P-glycoprotein, reduce the volume of distribution and/or clearance of digoxin, doubling its serum level
One negatively affects the other’s metabolismOne negatively affects the other’s oxidative metabolism by inducing CYP enzyme activityCarbamazepine induces CYP 2C9 and CYP 3A4 activity, which stimulates warfarin biotransformation, decreases its half-life, and lowers its serum concentration
 One negatively affects the other’s oxidative metabolism by inhibiting CYP enzyme activityKetoconazole inhibits CYP 3A4 activity, which inhibits terfenadine metabolism, resulting in serum terfenadine levels 32 to 100 times normal
 One inhibits hydroxylation of the other’s toxic metabolites, inhibiting their clearanceCombination of carbamazepine and valproate
One negatively affects the other’s elimination Lithium plus hydrochlorothiazide or an NSAID (both impair lithium excretion)

This addition to the equation explains how inter-individual variability can shift the dose-response curve to produce a greater or lesser effect than that which would be expected in the “usual” patient taking the prescribed dosage.

Inter-individual variance. The metabolism of dextromethorphan illustrates the effect of inter-individual variance. After a single dose, about 93% of Caucasians develop relatively lower dextromethorphan:dextrophan ratios, and about 7% develop relatively higher ratios. This difference defines patients who are pharmacogenetically CYP 2D6 extensive metabolizers versus those who are not.

Similarly, drugs sometimes cause biological variance, which predisposes to a drug-drug interaction. For example, the literature is replete with case reports and case series reporting that a substantial CYP 2D6 inhibitor—such as fluoxetine—blocks the metabolism of drugs that are principally metabolized by CYP 2D6. If the drug being metabolized has a narrow therapeutic index—such as amitriptyline—the resultant increase in its serum level can cause serious cardio and neurotoxicity, including arrhythmias, delirium, seizures, coma, and death.12

In such cases, a CYP 2D6 inhibitor converts the phenotype from a CYP 2D6 extensive metabolizer into a CYP 2D6 poor metabolizer. Hence, the clinician must consider how a specific patient may differ from the usual patient when selecting and dosing a drug. The difference may be genetic or acquired, as in this example.

Table 4

RISK FACTORS FOR POLYPHARMACY

Psychiatric disordersMedications being taken
SchizophreniaCardiovascular agents
Bipolar disorderAntipsychotics
DepressionMood stabilizers
Borderline and other personality disordersAntidepressants
Substance abuse (including tobacco habituation)Self-medication with aspirin
Neurologic disordersDemographic variables
Mental retardationAge 65 or older
DementiaEthnicity (Caucasian, African-American)
Chronic pain, facial painFemale gender
Headache (including migraine)Psychosocial variables
InsomniaLower socioeconomic status
EpilepsyInner-city residence
Medical disordersLower level of education
Chronic diseases, multiple diseasesUnemployment
ObesitySelf-medication
DiabetesConcealed drug use
Chronic hypertension 
Coronary artery disease 

The following equation explains how dose is related to drug concentration, which takes into account the drug’s pharmacokinetics:

drug concentration = dosing rate (mg/day) ÷ clearance (ml/min)

In other words, the concentration achieved in a specific patient is determined by the dosage relative to the patient’s ability to clear the drug from the body.

Consequences, prevalence of polypharmacy

Polypharmacy increases patients’ risk for many ill effects, including incidence and severity of adverse events, drug-drug interactions, medication errors, hospitalizations, morbidity, mortality, and direct and indirect costs. At least 12 reports and studies have been published showing the association between polypharmacy and death,2,13-23 and in some of these reports the association is present even after controlling for underlying diseases.

The prevalence of polypharmacy varies by country and population. In Denmark, for example, the prevalence of polypharmacy is approximately 1.2%,6 compared with approximately 7% in the United States.24 Nearly one-half (46%) of all elderly persons admitted to U.S. hospitals may be taking seven or more medications.25 Polypharmacy is especially problematic in patients age 65 and older (Box 2),26-31 in whom the top five preventable threats to health are congestive heart failure, breast cancer, hypertension, pneumonia, and adverse drug events.32 Although older persons make up less than 15% of the population, they take the greatest number and quantity of medications, purchase 40% of all nonprescription medications, and use 33% of all retail prescriptions.30

Box 2

POLYPHARMACY RISKS IN PATIENTS AGE 65 AND OLDER

  • 14% of older patients prescribed psychotropics experience a hip fracture, accounting for 32,000 annual hip fractures in the United States.26
  • 28% of older patients’ hospitalizations are due to adverse events or non-adherence to drug therapy.27
  • 35% of older patients taking three or more prescription medications at hospital discharge are re-hospitalized within 6 months. Problems with medications lead to 6.4% of these re-admissions.28
  • Among older drivers, taking a psychoactive drug multiplies the risk of a motor vehicle accident involving injuries by 1.5 to 5.5 times. The greater the dosage, the greater the risk.29
  • Hospital admissions related to adverse events from medications in older patients cost $20 billion annually (excluding indirect costs).30
  • Morbidity and mortality related to drug therapy in ambulatory patients in the United States costs $76.6 billion annually.31
 

 

Psychiatric disorders including schizophrenia, bipolar disorder, depression, personality disorders, and substance abuse place patients at higher risk for polypharmacy, as do certain demographic, psychosocial, medication, medical, and neurologic factors (Table 4). Other factors that increase the risk for polypharmacy include:

  • institutional factors (recent hospitalization, admission to a surgical ward, nursing home placement, home health care, increased number of pharmacies used, increased number of clinics attended, client-centered psychiatric treatment compared with non-client-centered psychiatric treatment)
  • provider factors (visit to a physician, treatment by general practitioners compared with specialists, increased number of providers, undocumented rationale or diagnosis supporting multiple medication use)
  • having medical insurance.

Steps to avoiding polypharmacy

By identifying polypharmacy’s risk factors, we may decrease its associated morbidity, mortality, and cost. Steps to follow while prescribing—as represented by the mnemonics SAIL33 and TIDE—may help you avoid polypharmacy’s negative consequences.

SAIL. Keep the drug regimen as simple as possible. Aim for once-daily or twice daily dosing. Try to simplify complex drug regimens by discontinuing any drug that does not achieve its defined therapeutic goal. For diseases and syndromes with less clear-cut causes, subtracting drugs from a complicated regimen may be more therapeutic than adding another drug. Try to treat multiple symptoms and syndromes with a single drug that may have multiple beneficial effects, rather than treating each symptom or syndrome with individual drugs.

Understand the potential adverse effects of each drug and potential drug-drug interactions. Whenever practical, choose drugs with broad rather than narrow therapeutic indices.

Each prescribed drug should have a clear indication and a well-defined therapeutic goal. Prescribe using evidence-based medicine as much as is practical.

List the name and dosage of each drug in the patient’s chart, and provide this information to the patient.33 Consider adopting computer data entry and feedback procedures, which have been shown to decrease polypharmacy34 and drug-drug interactions.35

TIDE. In the busy medical practice, writing a prescription signals to the patient that his or her time with the doctor is almost finished. Allow time to address medication issues.

Apply the understanding of individual variability, pharmacokinetics, and pharmacodynamics when prescribing. Review with the patient all prescription and nonprescription drugs and dietary supplements being taken.

Be careful to avoid potentially dangerous drug-drug interactions, especially those associated with serious adverse events such as torsades de pointes.

Educate patients regarding drug and non-drug treatments. Explain potential adverse effects of each drug and potential drug-drug interactions.

Related resources

Disclosure

Drs. Werder and Preskorn have served on the speakers bureau of, as consultants to, or as principal investigators for Abbott Laboratories, AstraZeneca Pharmaceuticals, Biovail Corp., Bristol-Meyers Squibb Co., Merck and Co., Eisai Inc., Eli Lilly and Co., GlaxoSmithKline, Hoffman-LaRoche, Janssen Pharmaceutica, Lundbeck, Novartis Pharmaceuticals Corp., Organon, Pfizer Inc., Solvay, Wyeth Pharmaceuticals, and Yamanouchi Pharmaceuticals Co., Ltd.

“Do no harm” is the first rule of medicine, yet 106,000 Americans die each year from properly prescribed and correctly taken medications.1 In some cases, the cause of death is known and can be attributed to a drug-drug interaction. The likelihood of death or hospitalization is directly proportional to the number of medications a patient is taking, even after controlling for underlying diseases.2

In psychiatry, it is not unusual for us to prescribe more than one psychotropic agent to manage a patient’s symptoms:

  • Patients with affective and psychotic disorders are commonly prescribed combinations of antipsychotics, mood stabilizers, antidepressants (often from more than one class), anxiolytics, antihistamines, and anticholinergics.
  • Patients with posttraumatic stress disorder may take selective serotonin reuptake inhibitors, buspirone, trazodone, antipsychotics, mood stabilizers, benzodiazepines, beta blockers, and opiates.
  • Multiple-drug regimens are used in treating other medical and psychiatric disorders, including chronic pain, fibromyalgia, chronic fatigue syndrome, sleep disorders, and epilepsy.

The greater the number of drugs used, the greater the likelihood that adverse events are emerging and are being treated, sometimes while being mistaken for patient psychopathology. As a prescriber, you are in a unique position to recognize and prevent interactions that can occur when patients are treated with two or more medications. This article defines polypharmacy, describes its consequences, prevalence, and risk factors, and offers an eight-step strategy with two mnemonics to help you avoid adverse events when prescribing multiple-drug regimens.

Box 1

POLYPHARMACY: MANY DRUGS, MANY DEFINITIONS

Poly, from the Greek word polus (many, much) and pharmacy, from the Greek word pharmakon (drug, poison) literally means many drugs or, alternatively, much poison.3 The word polypharmacy first appeared in the medical literature in 1959 in the New England Journal of Medicine4 and in the psychiatric literature in 1969 in an article citing its incidence at a state mental hospital.5

Many definitions have been used to describe and define polypharmacy, both qualitatively and quantitatively. Monotherapy is drug treatment with one drug. Sometimes treatment with two drugs is referred to as co-pharmacy, while treatment with three or more drugs is referred to as polypharmacy.Minor polypharmacy refers to treatment with two to four drugs, while major polypharmacyrefers to treatment with five or more drugs.6

What is polypharmacy?

Many definitions have been used to describe polypharmacy (Box 1).3-6 The most common definition is the use of five or more drugs at the same time in the same patient.7 Although polypharmacy often has a pejorative connotation, using five or more drugs may be therapeutic or contratherapeutic.

Therapeutic polypharmacy occurs, for example, when expert panels or researchers in carefully controlled clinical trials recommend using multiple medications to treat specific diseases. For example, the five-drug combination of isoniazid, rifampin, ethambutol, pyrazinamide, and pyridoxine is therapeutic in initial tuberculosis treatment. More is better in this case because four antibiotics are needed to prevent the development of multiple drug-resistant Mycobacterium tuberculosis, and adding pyridoxine prevents isoniazid-induced neurotoxicity. This example illustrates two prescribing principles:

  • using multiple drugs can help achieve an intended therapeutic goal
  • adding one drug can prevent a known side effect of another drug.

Another example is the therapeutic management of congestive heart failure, in which five drug classes—an angiotensin-converting enzyme (ACE) inhibitor, a diuretic, a digitalis glycoside, a beta blocker, and an aldosterone antagonist—are used in various combinations. All play a role in improving cardiac function and reducing morbidity and mortality.

Using combination drug therapy can also generate cost benefits, such as by adding a drug to delay or inhibit the metabolism of an expensive principal drug. For example, adding diltiazem—a cytochrome P450 (CYP) 3A4 inhibitor—to cyclosporine—which is metabolized by CYP 3A4 enzymes—reduces the dosage of cyclosporine needed to achieve a desired serum level, thereby reducing the cost of this drug. (Some have abandoned this strategy because of cyclosporine’s narrow therapeutic index.)

Contratherapeutic polypharmacy occurs when a patient taking multiple drugs experiences an unexpected or unintended adverse outcome.

Settings for polypharmacy

Polypharmacy occurs in five principal prescribing situations:

  • treatment of symptoms
  • treatment of multiple illnesses
  • treatment of phasic illnesses, such as many affective, anxiety, seizure, and neurodegenerative disorders
  • preventing or treating adverse effects of other drugs
  • attempting to accelerate the onset of action or augment the effects of a preceding drug.

As described above, diseases such as tuberculosis and congestive heart failure, with well-understood causes and pathophysiologies, are often treated with multiple therapeutic drug combinations. However, the causes of many psychiatric disorders and syndromes are less well-understood, which makes prescribing drug combinations more difficult. It may be that treating less well-understood diseases is a risk factor for contratherapeutic polypharmacy.

 

 

Most individuals who are prescribed five or more drugs are taking unique drug combinations.8 These heterogeneous regimens represent “an uncontrolled experiment,” with effects that cannot be predicted from studies in the literature.9Tables 1, 2, and 3 describe how contratherapeutic polypharmacy may occur with combinations of any number of drugs, whether five or more by the classic definition or only two. For example, contratherapeutic polypharmacy may occur when a patient is given the mood-stabilizing drugs valproate and carbamazepine (CBZ) at the same time.10 Here is why this combination may be dangerous:

  • Carbamazepine is oxidized by arene oxidase to CBZ 10,11-epoxide, which is hydrolyzed by epoxide hydrolase to CBZ 10,11-dihydroxide. The metabolite CBZ 10,11-epoxide has both therapeutic and toxic effects.
  • In monotherapy, the ratio of carbamazepine to CBZ 10,11-epoxide is 10:1, with CBZ 10,11-epoxide having a shorter half-life than carbamazepine.
  • However, when carbamazepine and valproate are taken as co-pharmacy, valproate blocks the hydrolysis of CBZ 10,11-epoxide by inhibiting epoxide hydrolase, so that the ratio of carbamazepine to CBZ 10,11-epoxide becomes 2:1. Higher concentrations of the epoxide metabolite contribute to neurotoxicity.

Table 1

POLYPHARMACY WITH TWO OR MORE MEDICATIONS

DescriptionExample
Two or more drugs from the same drug categoryTwo nonsteroidal anti-inflammatory drugs (NSAIDs), two ACE inhibitors, or two phenothiazines
Use of multiple medications across therapeutic classesUse of multiple CNS medications, as in multiple antidepressants, antipsychotics, or anticonvulsants
An inappropriate or unnecessary medication is prescribed to a patient taking other medicationInappropriate prescription due to relative or absolute contraindications Inappropriate prescription due to weak or no indication
Prescription of an exceedingly high dose to a patient taking other medicationThe maximum recommended dose may be functionally exceeded to a serious degree if a drug with a narrow therapeutic index (e.g., amitriptyline) is combined with one that blocks its metabolism (e.g., fluoxetine)
Two or more drugs sharing similar toxicitiesAnticholinergic toxicity due to combining a low-potency phenothiazine antipsychotic and a tertiary amine tricyclic antidepressant

Other examples of potentially dangerous drug combinations include those associated with torsades de pointes, which may occur with certain combinations of antihistamines, antidepressants, antipsychotics, antivirals, antibacterials, antifungals, antiarrhythmics, and promotility agents.

Drug-drug interactions

In a drug-drug interaction, the presence of one drug alters the nature, magnitude, or duration of the effect of a given dose of another drug; the interaction may be either therapeutic or adverse, depending on the desired effect. A drug-drug interaction may be intended or unintended and is determined by pharmacokinetics and pharmacodynamics rather than by therapeutic class.

Most available drug information describes the effects of individual drugs used alone (monopharmacy). Information on how one drug interacts with another (co-pharmacy) is more difficult to come by. A recent literature search using broad criteria for drug-drug interactions uncovered 4,277 indexed articles. Another search, this time using narrow criteria, produced only 316 articles, suggesting that systematic studies regarding drug-drug interactions are few.

Table 2

HOW PHARMACODYNAMICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

MechanismExamples
One drug has a mechanism of action directly opposing the mechanism of action of a co-prescribed drugBromocriptine and prochlorperazine in treating a patient with parkinsonism and nausea
Levidopa/carbidopa and risperidone in treating a patient with parkinsonism and psychosis
Venlafaxine and atenolol in treating a patient with depression and hypertension
One drug has an action that increases the potential for an adverse event of a co-prescribed drugOrthostatic hypotension and syncope when an ACE inhibitor is added to a diuretic
Orthostatic hypotension and syncope when risperidone, because of its action as an alpha-1 adrenergic blocker, is added to a diuretic
Narcosis and respiratory failure when parenteral fentanyl is added to oral meperidine
Neurotoxicity (absence status epilepticus) when valproate is added to clonazepam in children with absence seizures

However, if you understand the pharmacodynamics and pharmacokinetics that rule co-pharmacy, then you can apply this knowledge to more complex drug-drug interactions involving contratherapeutic polypharmacy.

How drug effects are determined. The nature and magnitude of a drug’s effect are determined by its site of action and its binding affinity, concentration, and action at that site.11 This relationship can be represented by the formula:

effect = potency at the site of action × concentration at the site of action

Potency at the site of action is determined by the binding affinity for the drug and the degree to which the receptor is stimulated or blocked, thus activating or inhibiting transmembrane and intracellular messengers (pharmacodynamics). Concentration at the site of action is determined by absorption, metabolism, distribution, and elimination (pharmacokinetics). Thus, the above model can be represented mathematically by:

effect = pharmacodynamics × pharmacokinetics

These factors determine a drug’s usual effect in the usual patient on the usual dosage, which is the goal of most clinical trials. However, all patients are not “usual,” because of inter-individual differences due to genetics, gender, age, environment, social habits such as smoking, intercurrent diseases affecting organ function, and concomitant drug therapy. Thus, when we take these factors into account, the first mathematical equation becomes:

 

 

effect = potency at the site of action × concentration at site of action × inter-individual variance

In other words, the clinical response equals the drug’s potency at the site of action times the drug’s concentration at the site of action times the patient’s underlying biology. Likewise, when we consider variability among patients, the second equation becomes:

effect = pharmacodynamics × pharmacokinetics × inter-individual variance

Table 3

HOW PHARMACOKINETICS MAY CAUSE ADVERSE DRUG-DRUG EVENTS

Mechanism of interaction of two or more drugsTwo or more drugs interact where …Examples
One negatively affects the other’s absorption Use of tetracycline with substances containing calcium
One negatively affects the other’s distribution Amiodarone and quinidine, by inhibiting P-glycoprotein, reduce the volume of distribution and/or clearance of digoxin, doubling its serum level
One negatively affects the other’s metabolismOne negatively affects the other’s oxidative metabolism by inducing CYP enzyme activityCarbamazepine induces CYP 2C9 and CYP 3A4 activity, which stimulates warfarin biotransformation, decreases its half-life, and lowers its serum concentration
 One negatively affects the other’s oxidative metabolism by inhibiting CYP enzyme activityKetoconazole inhibits CYP 3A4 activity, which inhibits terfenadine metabolism, resulting in serum terfenadine levels 32 to 100 times normal
 One inhibits hydroxylation of the other’s toxic metabolites, inhibiting their clearanceCombination of carbamazepine and valproate
One negatively affects the other’s elimination Lithium plus hydrochlorothiazide or an NSAID (both impair lithium excretion)

This addition to the equation explains how inter-individual variability can shift the dose-response curve to produce a greater or lesser effect than that which would be expected in the “usual” patient taking the prescribed dosage.

Inter-individual variance. The metabolism of dextromethorphan illustrates the effect of inter-individual variance. After a single dose, about 93% of Caucasians develop relatively lower dextromethorphan:dextrophan ratios, and about 7% develop relatively higher ratios. This difference defines patients who are pharmacogenetically CYP 2D6 extensive metabolizers versus those who are not.

Similarly, drugs sometimes cause biological variance, which predisposes to a drug-drug interaction. For example, the literature is replete with case reports and case series reporting that a substantial CYP 2D6 inhibitor—such as fluoxetine—blocks the metabolism of drugs that are principally metabolized by CYP 2D6. If the drug being metabolized has a narrow therapeutic index—such as amitriptyline—the resultant increase in its serum level can cause serious cardio and neurotoxicity, including arrhythmias, delirium, seizures, coma, and death.12

In such cases, a CYP 2D6 inhibitor converts the phenotype from a CYP 2D6 extensive metabolizer into a CYP 2D6 poor metabolizer. Hence, the clinician must consider how a specific patient may differ from the usual patient when selecting and dosing a drug. The difference may be genetic or acquired, as in this example.

Table 4

RISK FACTORS FOR POLYPHARMACY

Psychiatric disordersMedications being taken
SchizophreniaCardiovascular agents
Bipolar disorderAntipsychotics
DepressionMood stabilizers
Borderline and other personality disordersAntidepressants
Substance abuse (including tobacco habituation)Self-medication with aspirin
Neurologic disordersDemographic variables
Mental retardationAge 65 or older
DementiaEthnicity (Caucasian, African-American)
Chronic pain, facial painFemale gender
Headache (including migraine)Psychosocial variables
InsomniaLower socioeconomic status
EpilepsyInner-city residence
Medical disordersLower level of education
Chronic diseases, multiple diseasesUnemployment
ObesitySelf-medication
DiabetesConcealed drug use
Chronic hypertension 
Coronary artery disease 

The following equation explains how dose is related to drug concentration, which takes into account the drug’s pharmacokinetics:

drug concentration = dosing rate (mg/day) ÷ clearance (ml/min)

In other words, the concentration achieved in a specific patient is determined by the dosage relative to the patient’s ability to clear the drug from the body.

Consequences, prevalence of polypharmacy

Polypharmacy increases patients’ risk for many ill effects, including incidence and severity of adverse events, drug-drug interactions, medication errors, hospitalizations, morbidity, mortality, and direct and indirect costs. At least 12 reports and studies have been published showing the association between polypharmacy and death,2,13-23 and in some of these reports the association is present even after controlling for underlying diseases.

The prevalence of polypharmacy varies by country and population. In Denmark, for example, the prevalence of polypharmacy is approximately 1.2%,6 compared with approximately 7% in the United States.24 Nearly one-half (46%) of all elderly persons admitted to U.S. hospitals may be taking seven or more medications.25 Polypharmacy is especially problematic in patients age 65 and older (Box 2),26-31 in whom the top five preventable threats to health are congestive heart failure, breast cancer, hypertension, pneumonia, and adverse drug events.32 Although older persons make up less than 15% of the population, they take the greatest number and quantity of medications, purchase 40% of all nonprescription medications, and use 33% of all retail prescriptions.30

Box 2

POLYPHARMACY RISKS IN PATIENTS AGE 65 AND OLDER

  • 14% of older patients prescribed psychotropics experience a hip fracture, accounting for 32,000 annual hip fractures in the United States.26
  • 28% of older patients’ hospitalizations are due to adverse events or non-adherence to drug therapy.27
  • 35% of older patients taking three or more prescription medications at hospital discharge are re-hospitalized within 6 months. Problems with medications lead to 6.4% of these re-admissions.28
  • Among older drivers, taking a psychoactive drug multiplies the risk of a motor vehicle accident involving injuries by 1.5 to 5.5 times. The greater the dosage, the greater the risk.29
  • Hospital admissions related to adverse events from medications in older patients cost $20 billion annually (excluding indirect costs).30
  • Morbidity and mortality related to drug therapy in ambulatory patients in the United States costs $76.6 billion annually.31
 

 

Psychiatric disorders including schizophrenia, bipolar disorder, depression, personality disorders, and substance abuse place patients at higher risk for polypharmacy, as do certain demographic, psychosocial, medication, medical, and neurologic factors (Table 4). Other factors that increase the risk for polypharmacy include:

  • institutional factors (recent hospitalization, admission to a surgical ward, nursing home placement, home health care, increased number of pharmacies used, increased number of clinics attended, client-centered psychiatric treatment compared with non-client-centered psychiatric treatment)
  • provider factors (visit to a physician, treatment by general practitioners compared with specialists, increased number of providers, undocumented rationale or diagnosis supporting multiple medication use)
  • having medical insurance.

Steps to avoiding polypharmacy

By identifying polypharmacy’s risk factors, we may decrease its associated morbidity, mortality, and cost. Steps to follow while prescribing—as represented by the mnemonics SAIL33 and TIDE—may help you avoid polypharmacy’s negative consequences.

SAIL. Keep the drug regimen as simple as possible. Aim for once-daily or twice daily dosing. Try to simplify complex drug regimens by discontinuing any drug that does not achieve its defined therapeutic goal. For diseases and syndromes with less clear-cut causes, subtracting drugs from a complicated regimen may be more therapeutic than adding another drug. Try to treat multiple symptoms and syndromes with a single drug that may have multiple beneficial effects, rather than treating each symptom or syndrome with individual drugs.

Understand the potential adverse effects of each drug and potential drug-drug interactions. Whenever practical, choose drugs with broad rather than narrow therapeutic indices.

Each prescribed drug should have a clear indication and a well-defined therapeutic goal. Prescribe using evidence-based medicine as much as is practical.

List the name and dosage of each drug in the patient’s chart, and provide this information to the patient.33 Consider adopting computer data entry and feedback procedures, which have been shown to decrease polypharmacy34 and drug-drug interactions.35

TIDE. In the busy medical practice, writing a prescription signals to the patient that his or her time with the doctor is almost finished. Allow time to address medication issues.

Apply the understanding of individual variability, pharmacokinetics, and pharmacodynamics when prescribing. Review with the patient all prescription and nonprescription drugs and dietary supplements being taken.

Be careful to avoid potentially dangerous drug-drug interactions, especially those associated with serious adverse events such as torsades de pointes.

Educate patients regarding drug and non-drug treatments. Explain potential adverse effects of each drug and potential drug-drug interactions.

Related resources

Disclosure

Drs. Werder and Preskorn have served on the speakers bureau of, as consultants to, or as principal investigators for Abbott Laboratories, AstraZeneca Pharmaceuticals, Biovail Corp., Bristol-Meyers Squibb Co., Merck and Co., Eisai Inc., Eli Lilly and Co., GlaxoSmithKline, Hoffman-LaRoche, Janssen Pharmaceutica, Lundbeck, Novartis Pharmaceuticals Corp., Organon, Pfizer Inc., Solvay, Wyeth Pharmaceuticals, and Yamanouchi Pharmaceuticals Co., Ltd.

References

1. Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 1998;279(15):1200-5.

2. Incalzi RA, Gemma A, Capparella O, et al. Predicting mortality and length of stay of geriatric patients in an acute care general hospital. J Gerontol 1992;47(2):M35-9.

3. Berube MS, Neely DJ, DeVinne PB. American Heritage Dictionary. (2nd College ed). Boston: Houghton Mifflin Co, 1982.

4. Friend DG. Polypharmacy: multiple-ingredient and shotgun prescriptions. N Engl J Med 1959;260(20):1015-8.

5. Sheppard C, Collins L, Fiorentino D, Fracchia J, Merlis S. Polypharmacy in psychiatric treatment. I. Incidence at a state hospital. Curr Ther Res Clin Exp 1969;(12):765-74.

6. Bjerrum L, Rosholm JU, Hallas J, Kragstrup J. Methods for estimating the occurrence of polypharmacy by means of a prescription database. Eur J Clin Pharmacol 1997;53(1):7-11.

7. Werder SF. Polypharmacy: definitions and risk factors (grand rounds). University of Kansas School of Medicine-Wichita, Department of Psychiatry and Behavioral Sciences. Via Christi Regional Medical Center, St. Joseph Campus: Dec 12, 2000.

8. Bjerrum L, Sogaard J, Hallas J, Kragstrup J. Polypharmacy: correlations with sex, age and drug regimen. A prescription database study. Eur J Clin Pharmacol 1998;54(3):197-202.

9. Bjerrum L. Pharmacoepidemiological Studies of Polypharmacy: Methodological issues, population estimates, and influence of practice patterns (PhD thesis). Odense University Faculty of Health Sciences, Department of clinical pharmacology and research unit of general practice. Denmark; 1998. Available at http://www.sdu.dk/health/IPH/genpract/staff/lbjerrum/PHD/PHD.HTM. Accessed Jan. 9, 2003.

10. Wilder BJ. Pharmacokinetics of valproate and carbamazepine. J Clin Psychopharmacol 1992;12(1 suppl):64S-68S.

11. Preskorn SH. The rational basis for the development and use of newer antidepressants. In: Outpatient management of depression: a guide for the practitioner (2nd ed). Caddo, OK: Professional Publications, Inc; 1999;57-103.

12. Vaughan DA. Interaction of fluoxetine with tricyclic antidepressants. Am J Psychiatry 1988;145(11):1478.-

13. Meeker JE, Reynolds PC. Postmortem tissue methamphetamine concentrations following selegiline administration. J Anal Toxicol 1990;14(5):330-1.

14. Sallee FR, DeVane CL, Ferrell RE. Fluoxetine-related death in a child with cytochrome P-450 2D6 genetic deficiency. J Child Adolesc Psychopharmacol 2000;10(1):27-34.

15. Ellis RJ, Mayo MS, Bodensteiner DM. Ciprofloxacin-warfarin coagulopathy: a case series. Am J Hematol 2000;63(1):28-31.

16. Konig SA, Siemes H, Blaker F, et al. Severe hepatotoxicity during valproate therapy: an update and report of eight new fatalities. Epilepsia 1994;35(5):1005-15.

17. Fattinger K, Roos M, Vergeres P, et al. Epidemiology of drug exposure and adverse drug reactions in two Swiss departments of internal medicine. Br J Clin Pharmacol 2000;49(2):158-67.

18. Ebbesen J, Buajordet I, Erikssen J, Svaar H, Brors O, Hilberg T. Drugs as a cause of death. A prospective quality assurance project in a department of medicine (Norwegian). Tidsskr Nor Laegeforen 1995;115(19):2369-72.

19. Alarcon T, Barcena A, Gonzalez-Montalvo JI, Penalosa C, Salgado A. Factors predictive of outcome on admission to an acute geriatric ward. Age Ageing 1999;28(5):429-32.

20. Smith NK, Albazzaz MK. A prospective study of urinary retention and risk of death after proximal femoral fracture. Age Ageing 1996;25(2):150-4.

21. Pulska T, Pahkala K, Laippala P, Kivela SL. Six-year survival of depressed elderly Finns: a community study. Int J Geriatr Psychiatry 1997;12(9):942-50.

22. Waddington JL, Youssef HA, Kinsella A. Mortality in schizophrenia. Antipsychotic polypharmacy and absence of adjunctive anticholinergics over the course of a 10-year prospective study. Br J Psychiatry 1998;173:325-9.

23. Burns R, Nichols LO, Graney MJ, Cloar FT. Impact of continued geriatric outpatient management on health outcomes of older veterans. Arch Intern Med 1995;155(12):1313-8.

24. Kaufman DW, Kelly JP, Rosenberg L, Anderson TE, Mitchell AA. Recent patterns of medication use in the ambulatory adult population of the United States: the Slone survey. JAMA 2002;16;287(3):337-44.

25. Flaherty JH, Perry HM, 3rd, Lynchard GS, Morley JE. Polypharmacy and hospitalization among older home care patients. J Gerontol A Biol Sci Med Sci 2000;55(10):M554-9.

26. Ray WA, Griffin MR, Schaffner W, Baugh DK, Melton LJ, 3rd. Psychotropic drug use and the risk of hip fracture. N Engl J Med 1987;316(7):363-9.

27. Col N, Fanale JE, Kronholm P. The role of medication noncompliance and adverse drug reactions in hospitalizations of the elderly. Arch Intern Med 1990;150(4):841-5.

28. Bero LA, Lipton HL, Bird JA. Characterization of geriatric drug-related hospital readmissions. Med Care 1991;29(10):989-1003.

29. Ray WA, Fought RL, Decker MD. Psychoactive drugs and the risk of injurious motor vehicle crashes in elderly drivers. Am J Epidemiol 1992;136(7):873-83.

30. Prescription drugs and the elderly. Publication AO/HEHS-95-152. Washington, DC: U.S. General Accounting Office, July 1995.

31. Johnson JA, Bootman JL. Drug-related morbidity and mortality. A cost-of-illness model. Arch Intern Med 1995;155(18):1949-56.

32. Fink A, Siu AL, Brook RH, Park RE, Solomon DH. Assuring the quality of health care for older persons. An expert panel’s priorities. JAMA 1987;258(14):1905-8.

33. Lee DR. Polypharmacy: a case report and new protocol for management. J Am Board Fam Pract 1998;11(2):140-4.

34. Hamdy RC, Moore SW, Whalen K, et al. Reducing polypharmacy in extended care. South Med J 1995;88(5):534-8.

35. Haumschild MJ, Ward ES, Bishop JM, Haumschild MS. Pharmacy-based computer system for monitoring and reporting drug interactions. Am J Hosp Pharm 1987;44(2):345-8.

References

1. Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 1998;279(15):1200-5.

2. Incalzi RA, Gemma A, Capparella O, et al. Predicting mortality and length of stay of geriatric patients in an acute care general hospital. J Gerontol 1992;47(2):M35-9.

3. Berube MS, Neely DJ, DeVinne PB. American Heritage Dictionary. (2nd College ed). Boston: Houghton Mifflin Co, 1982.

4. Friend DG. Polypharmacy: multiple-ingredient and shotgun prescriptions. N Engl J Med 1959;260(20):1015-8.

5. Sheppard C, Collins L, Fiorentino D, Fracchia J, Merlis S. Polypharmacy in psychiatric treatment. I. Incidence at a state hospital. Curr Ther Res Clin Exp 1969;(12):765-74.

6. Bjerrum L, Rosholm JU, Hallas J, Kragstrup J. Methods for estimating the occurrence of polypharmacy by means of a prescription database. Eur J Clin Pharmacol 1997;53(1):7-11.

7. Werder SF. Polypharmacy: definitions and risk factors (grand rounds). University of Kansas School of Medicine-Wichita, Department of Psychiatry and Behavioral Sciences. Via Christi Regional Medical Center, St. Joseph Campus: Dec 12, 2000.

8. Bjerrum L, Sogaard J, Hallas J, Kragstrup J. Polypharmacy: correlations with sex, age and drug regimen. A prescription database study. Eur J Clin Pharmacol 1998;54(3):197-202.

9. Bjerrum L. Pharmacoepidemiological Studies of Polypharmacy: Methodological issues, population estimates, and influence of practice patterns (PhD thesis). Odense University Faculty of Health Sciences, Department of clinical pharmacology and research unit of general practice. Denmark; 1998. Available at http://www.sdu.dk/health/IPH/genpract/staff/lbjerrum/PHD/PHD.HTM. Accessed Jan. 9, 2003.

10. Wilder BJ. Pharmacokinetics of valproate and carbamazepine. J Clin Psychopharmacol 1992;12(1 suppl):64S-68S.

11. Preskorn SH. The rational basis for the development and use of newer antidepressants. In: Outpatient management of depression: a guide for the practitioner (2nd ed). Caddo, OK: Professional Publications, Inc; 1999;57-103.

12. Vaughan DA. Interaction of fluoxetine with tricyclic antidepressants. Am J Psychiatry 1988;145(11):1478.-

13. Meeker JE, Reynolds PC. Postmortem tissue methamphetamine concentrations following selegiline administration. J Anal Toxicol 1990;14(5):330-1.

14. Sallee FR, DeVane CL, Ferrell RE. Fluoxetine-related death in a child with cytochrome P-450 2D6 genetic deficiency. J Child Adolesc Psychopharmacol 2000;10(1):27-34.

15. Ellis RJ, Mayo MS, Bodensteiner DM. Ciprofloxacin-warfarin coagulopathy: a case series. Am J Hematol 2000;63(1):28-31.

16. Konig SA, Siemes H, Blaker F, et al. Severe hepatotoxicity during valproate therapy: an update and report of eight new fatalities. Epilepsia 1994;35(5):1005-15.

17. Fattinger K, Roos M, Vergeres P, et al. Epidemiology of drug exposure and adverse drug reactions in two Swiss departments of internal medicine. Br J Clin Pharmacol 2000;49(2):158-67.

18. Ebbesen J, Buajordet I, Erikssen J, Svaar H, Brors O, Hilberg T. Drugs as a cause of death. A prospective quality assurance project in a department of medicine (Norwegian). Tidsskr Nor Laegeforen 1995;115(19):2369-72.

19. Alarcon T, Barcena A, Gonzalez-Montalvo JI, Penalosa C, Salgado A. Factors predictive of outcome on admission to an acute geriatric ward. Age Ageing 1999;28(5):429-32.

20. Smith NK, Albazzaz MK. A prospective study of urinary retention and risk of death after proximal femoral fracture. Age Ageing 1996;25(2):150-4.

21. Pulska T, Pahkala K, Laippala P, Kivela SL. Six-year survival of depressed elderly Finns: a community study. Int J Geriatr Psychiatry 1997;12(9):942-50.

22. Waddington JL, Youssef HA, Kinsella A. Mortality in schizophrenia. Antipsychotic polypharmacy and absence of adjunctive anticholinergics over the course of a 10-year prospective study. Br J Psychiatry 1998;173:325-9.

23. Burns R, Nichols LO, Graney MJ, Cloar FT. Impact of continued geriatric outpatient management on health outcomes of older veterans. Arch Intern Med 1995;155(12):1313-8.

24. Kaufman DW, Kelly JP, Rosenberg L, Anderson TE, Mitchell AA. Recent patterns of medication use in the ambulatory adult population of the United States: the Slone survey. JAMA 2002;16;287(3):337-44.

25. Flaherty JH, Perry HM, 3rd, Lynchard GS, Morley JE. Polypharmacy and hospitalization among older home care patients. J Gerontol A Biol Sci Med Sci 2000;55(10):M554-9.

26. Ray WA, Griffin MR, Schaffner W, Baugh DK, Melton LJ, 3rd. Psychotropic drug use and the risk of hip fracture. N Engl J Med 1987;316(7):363-9.

27. Col N, Fanale JE, Kronholm P. The role of medication noncompliance and adverse drug reactions in hospitalizations of the elderly. Arch Intern Med 1990;150(4):841-5.

28. Bero LA, Lipton HL, Bird JA. Characterization of geriatric drug-related hospital readmissions. Med Care 1991;29(10):989-1003.

29. Ray WA, Fought RL, Decker MD. Psychoactive drugs and the risk of injurious motor vehicle crashes in elderly drivers. Am J Epidemiol 1992;136(7):873-83.

30. Prescription drugs and the elderly. Publication AO/HEHS-95-152. Washington, DC: U.S. General Accounting Office, July 1995.

31. Johnson JA, Bootman JL. Drug-related morbidity and mortality. A cost-of-illness model. Arch Intern Med 1995;155(18):1949-56.

32. Fink A, Siu AL, Brook RH, Park RE, Solomon DH. Assuring the quality of health care for older persons. An expert panel’s priorities. JAMA 1987;258(14):1905-8.

33. Lee DR. Polypharmacy: a case report and new protocol for management. J Am Board Fam Pract 1998;11(2):140-4.

34. Hamdy RC, Moore SW, Whalen K, et al. Reducing polypharmacy in extended care. South Med J 1995;88(5):534-8.

35. Haumschild MJ, Ward ES, Bishop JM, Haumschild MS. Pharmacy-based computer system for monitoring and reporting drug interactions. Am J Hosp Pharm 1987;44(2):345-8.

Issue
Current Psychiatry - 02(02)
Issue
Current Psychiatry - 02(02)
Page Number
24-36
Page Number
24-36
Publications
Publications
Topics
Article Type
Display Headline
Managing polypharmacy: Walking the fine line between help and harm
Display Headline
Managing polypharmacy: Walking the fine line between help and harm
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media