User login
CDK inhibitor proves active against AML, ALL
PHILADELPHIA—Preclinical research suggests a cyclin-dependent kinase (CDK) inhibitor is active against acute leukemias, particularly those with mixed-lineage leukemia rearrangements (MLL-r).
CYC065 selectively inhibits CDK2, which drives cell-cycle transition and activates major DNA double-strand break repair pathways; CDK5, which drives metastatic spread; and CDK9, which regulates the transcription of genes important for the proliferation and survival of malignant cells.
Experiments have shown that CYC065 exhibits activity against acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL), with and without MLL-r.
Daniella Zheleva, PhD, and her colleagues described these experiments in a poster at the AACR Annual Meeting 2015 (abstract 1650). All of the researchers are employees of Cyclacel Ltd., the company developing CYC065.
The researchers tested CYC065 in a panel of AML cell lines with wild-type MLL (HEL, HL60, Kasumi-1, KG-1, OCI-AML5, and PL21) and MLL-r (EOL-1, ML-2, MOLM-13, MV4-11, Nomo-1, OCI-AML2, and THP-1).
They found that MLL-r cell lines were “highly sensitive” to CYC065, but the sensitivity of cell lines with wild-type MLL correlated with the level of Bcl-2 family proteins. In the wild-type cell lines, IC50/70/90 values were correlated with BclXL and inversely correlated with Bak.
Six-hour pulse treatment of CYC065 at 0.5 µM to 1 µM was sufficient to cause 90% or greater cell death in sensitive cell lines. And cell lines with reduced sensitivity to the drug could be targeted by exposure to 10-hour pulse treatments of CYC065, or to CYC065 in combination with short pulses of Bcl-2 inhibitors.
The researchers observed “potent antitumor activity” when they administered CYC065 in AML xenograft models.
In an EOL-1 model, the median tumor growth inhibition on day 19 was 97% for mice that received CYC065 at 40 mg/kg (every day on days 1-5 and 8-12), 95% for mice that received CYC065 at 20 mg/kg every day on days 1-5 and 8-12), and 41% for mice that received cytarabine at 100 mg/kg (every day on days 1-5).
In the HL60 model, the median tumor growth inhibition on day 11 was 90% for mice that received CYC065 at 70 mg/kg (every day on days 1-5 and 8-12). And 2 mice achieved a complete response to treatment.
The researchers also found that CYC065 synergizes with cytarabine, particularly when CYC065 is given first. In fact, the combination could overcome the cytarabine resistance observed in the MV4-11 cell line.
CYC065 was “strongly synergistic” with Bcl2/BclXL inhibitors as well, the researchers said. CYC065 synergized with ABT-199, ABT-263, and ABT-737 in both AML cell lines (THP-1 and HEL) and ALL cell lines (Jurkat and SEM).
The researchers said the potent in vitro and in vivo activity of CYC065 and the ability to combine the drug with other antileukemic agents suggest that it may have therapeutic potential in AML and ALL.
PHILADELPHIA—Preclinical research suggests a cyclin-dependent kinase (CDK) inhibitor is active against acute leukemias, particularly those with mixed-lineage leukemia rearrangements (MLL-r).
CYC065 selectively inhibits CDK2, which drives cell-cycle transition and activates major DNA double-strand break repair pathways; CDK5, which drives metastatic spread; and CDK9, which regulates the transcription of genes important for the proliferation and survival of malignant cells.
Experiments have shown that CYC065 exhibits activity against acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL), with and without MLL-r.
Daniella Zheleva, PhD, and her colleagues described these experiments in a poster at the AACR Annual Meeting 2015 (abstract 1650). All of the researchers are employees of Cyclacel Ltd., the company developing CYC065.
The researchers tested CYC065 in a panel of AML cell lines with wild-type MLL (HEL, HL60, Kasumi-1, KG-1, OCI-AML5, and PL21) and MLL-r (EOL-1, ML-2, MOLM-13, MV4-11, Nomo-1, OCI-AML2, and THP-1).
They found that MLL-r cell lines were “highly sensitive” to CYC065, but the sensitivity of cell lines with wild-type MLL correlated with the level of Bcl-2 family proteins. In the wild-type cell lines, IC50/70/90 values were correlated with BclXL and inversely correlated with Bak.
Six-hour pulse treatment of CYC065 at 0.5 µM to 1 µM was sufficient to cause 90% or greater cell death in sensitive cell lines. And cell lines with reduced sensitivity to the drug could be targeted by exposure to 10-hour pulse treatments of CYC065, or to CYC065 in combination with short pulses of Bcl-2 inhibitors.
The researchers observed “potent antitumor activity” when they administered CYC065 in AML xenograft models.
In an EOL-1 model, the median tumor growth inhibition on day 19 was 97% for mice that received CYC065 at 40 mg/kg (every day on days 1-5 and 8-12), 95% for mice that received CYC065 at 20 mg/kg every day on days 1-5 and 8-12), and 41% for mice that received cytarabine at 100 mg/kg (every day on days 1-5).
In the HL60 model, the median tumor growth inhibition on day 11 was 90% for mice that received CYC065 at 70 mg/kg (every day on days 1-5 and 8-12). And 2 mice achieved a complete response to treatment.
The researchers also found that CYC065 synergizes with cytarabine, particularly when CYC065 is given first. In fact, the combination could overcome the cytarabine resistance observed in the MV4-11 cell line.
CYC065 was “strongly synergistic” with Bcl2/BclXL inhibitors as well, the researchers said. CYC065 synergized with ABT-199, ABT-263, and ABT-737 in both AML cell lines (THP-1 and HEL) and ALL cell lines (Jurkat and SEM).
The researchers said the potent in vitro and in vivo activity of CYC065 and the ability to combine the drug with other antileukemic agents suggest that it may have therapeutic potential in AML and ALL.
PHILADELPHIA—Preclinical research suggests a cyclin-dependent kinase (CDK) inhibitor is active against acute leukemias, particularly those with mixed-lineage leukemia rearrangements (MLL-r).
CYC065 selectively inhibits CDK2, which drives cell-cycle transition and activates major DNA double-strand break repair pathways; CDK5, which drives metastatic spread; and CDK9, which regulates the transcription of genes important for the proliferation and survival of malignant cells.
Experiments have shown that CYC065 exhibits activity against acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL), with and without MLL-r.
Daniella Zheleva, PhD, and her colleagues described these experiments in a poster at the AACR Annual Meeting 2015 (abstract 1650). All of the researchers are employees of Cyclacel Ltd., the company developing CYC065.
The researchers tested CYC065 in a panel of AML cell lines with wild-type MLL (HEL, HL60, Kasumi-1, KG-1, OCI-AML5, and PL21) and MLL-r (EOL-1, ML-2, MOLM-13, MV4-11, Nomo-1, OCI-AML2, and THP-1).
They found that MLL-r cell lines were “highly sensitive” to CYC065, but the sensitivity of cell lines with wild-type MLL correlated with the level of Bcl-2 family proteins. In the wild-type cell lines, IC50/70/90 values were correlated with BclXL and inversely correlated with Bak.
Six-hour pulse treatment of CYC065 at 0.5 µM to 1 µM was sufficient to cause 90% or greater cell death in sensitive cell lines. And cell lines with reduced sensitivity to the drug could be targeted by exposure to 10-hour pulse treatments of CYC065, or to CYC065 in combination with short pulses of Bcl-2 inhibitors.
The researchers observed “potent antitumor activity” when they administered CYC065 in AML xenograft models.
In an EOL-1 model, the median tumor growth inhibition on day 19 was 97% for mice that received CYC065 at 40 mg/kg (every day on days 1-5 and 8-12), 95% for mice that received CYC065 at 20 mg/kg every day on days 1-5 and 8-12), and 41% for mice that received cytarabine at 100 mg/kg (every day on days 1-5).
In the HL60 model, the median tumor growth inhibition on day 11 was 90% for mice that received CYC065 at 70 mg/kg (every day on days 1-5 and 8-12). And 2 mice achieved a complete response to treatment.
The researchers also found that CYC065 synergizes with cytarabine, particularly when CYC065 is given first. In fact, the combination could overcome the cytarabine resistance observed in the MV4-11 cell line.
CYC065 was “strongly synergistic” with Bcl2/BclXL inhibitors as well, the researchers said. CYC065 synergized with ABT-199, ABT-263, and ABT-737 in both AML cell lines (THP-1 and HEL) and ALL cell lines (Jurkat and SEM).
The researchers said the potent in vitro and in vivo activity of CYC065 and the ability to combine the drug with other antileukemic agents suggest that it may have therapeutic potential in AML and ALL.
Drug that fell short in prostate cancer could treat MM
PHILADELPHIA—A drug that has fallen short of expectations in clinical trials of prostate cancer may be effective for treating multiple myeloma (MM), according to research presented at the AACR Annual Meeting 2015.
The drug, tasquinimod, inhibits the function of S100A9, a pro-inflammatory protein that is elevated in MM, prostate cancer, and other malignancies.
Researchers found that tasquinimod can reduce tumor growth and improve survival in mouse models of MM. And these effects are associated with reduced angiogenesis in the bone marrow.
Cindy Lin, PhD, of the Wistar Institute in Philadelphia, Pennsylvania, and her colleagues presented these findings as abstract 1364.* The research was supported by Active Biotech and Ipsen, the companies developing tasquinimod.
Dr Lin noted that tasquinimod has already been tested in clinical trials of prostate cancer and initially appeared to be very effective. However, recent results from a phase 3 trial suggested the drug does not confer a favorable risk-benefit ratio for this population.
So Active Biotech and Ipsen decided to discontinue all trials of tasquinimod in prostate cancer. But the preclinical results observed in MM suggest tasquinimod may hold promise for treating these patients.
Activity in MM
Dr Lin said previous preclinical experiments revealed that myeloid-derived suppressor cells are involved in regulating MM progression, and these cells produce S100A9.
Tasquinimod is a quinoline-3-carboxamide derivative that binds to S100A9 and inhibits interaction with its receptors. So Dr Lin and her colleagues decided to investigate the antitumor effect of the drug in mouse models of MM.
The researchers initially tested tasquinimod in a syngeneic MM model, randomizing mice to treatment or control. Mice in the treatment group received tasquinimod at 30 mg/kg/day in their drinking water for 28 days.
Tasquinimod significantly improved survival in this model (P<0.005). All control mice died within 30 days of tumor inoculation, but about 40% of tasquinimod-treated mice were still alive more than 80 days out.
The researchers then tested tasquinimod in xenograft models of human MM. The drug significantly reduced tumor size in both H929 (P=0.0042) and RPMI-8226 models (P=0.0003). Treatment significantly improved survival in the H929 (P=0.0008) and RPMI-8226 models as well (P=0.0243).
Dr Lin said she and her colleagues did not see any side effects of treatment in any of the mice.
Investigating the mechanism
To determine if the antitumor effect of tasquinimod is, in fact, mediated through inhibition of S100A9, the researchers administered the drug to S100A9-knockout mice with MM. Tasquinimod did not improve survival in these mice, which suggests its anti-MM effects are mediated through S100A9 inhibition.
“To try and investigate some of the mechanisms of how survival is improved in tasquinimod-treated, tumor-bearing mice, we looked at a variety of different things, including angiogenesis,” Dr Lin said.
“We used CD31 immunohistochemistry to look at angiogenesis, and, in the untreated mice, we didn’t see a lot of staining. But in the tumor-bearing mice, there was a lot more staining [P=0.0231]. And when we gave the mice tasquinimod, angiogenesis was significantly decreased [P<0.0001].”
The researchers also looked at different angiogenic factors. And they found that, compared to control-treated mice with MM, tumor-bearing mice that received tasquinimod had a significant decrease in serum levels of VEGF, FGF2, tissue factor, and endoglin.
The team is now assessing the effects of S100A9 and tasquinimod on megakaryocytes and platelets, 2 of the major cell populations that promote angiogenesis.
*Information in the abstract differs from that presented at the meeting.
PHILADELPHIA—A drug that has fallen short of expectations in clinical trials of prostate cancer may be effective for treating multiple myeloma (MM), according to research presented at the AACR Annual Meeting 2015.
The drug, tasquinimod, inhibits the function of S100A9, a pro-inflammatory protein that is elevated in MM, prostate cancer, and other malignancies.
Researchers found that tasquinimod can reduce tumor growth and improve survival in mouse models of MM. And these effects are associated with reduced angiogenesis in the bone marrow.
Cindy Lin, PhD, of the Wistar Institute in Philadelphia, Pennsylvania, and her colleagues presented these findings as abstract 1364.* The research was supported by Active Biotech and Ipsen, the companies developing tasquinimod.
Dr Lin noted that tasquinimod has already been tested in clinical trials of prostate cancer and initially appeared to be very effective. However, recent results from a phase 3 trial suggested the drug does not confer a favorable risk-benefit ratio for this population.
So Active Biotech and Ipsen decided to discontinue all trials of tasquinimod in prostate cancer. But the preclinical results observed in MM suggest tasquinimod may hold promise for treating these patients.
Activity in MM
Dr Lin said previous preclinical experiments revealed that myeloid-derived suppressor cells are involved in regulating MM progression, and these cells produce S100A9.
Tasquinimod is a quinoline-3-carboxamide derivative that binds to S100A9 and inhibits interaction with its receptors. So Dr Lin and her colleagues decided to investigate the antitumor effect of the drug in mouse models of MM.
The researchers initially tested tasquinimod in a syngeneic MM model, randomizing mice to treatment or control. Mice in the treatment group received tasquinimod at 30 mg/kg/day in their drinking water for 28 days.
Tasquinimod significantly improved survival in this model (P<0.005). All control mice died within 30 days of tumor inoculation, but about 40% of tasquinimod-treated mice were still alive more than 80 days out.
The researchers then tested tasquinimod in xenograft models of human MM. The drug significantly reduced tumor size in both H929 (P=0.0042) and RPMI-8226 models (P=0.0003). Treatment significantly improved survival in the H929 (P=0.0008) and RPMI-8226 models as well (P=0.0243).
Dr Lin said she and her colleagues did not see any side effects of treatment in any of the mice.
Investigating the mechanism
To determine if the antitumor effect of tasquinimod is, in fact, mediated through inhibition of S100A9, the researchers administered the drug to S100A9-knockout mice with MM. Tasquinimod did not improve survival in these mice, which suggests its anti-MM effects are mediated through S100A9 inhibition.
“To try and investigate some of the mechanisms of how survival is improved in tasquinimod-treated, tumor-bearing mice, we looked at a variety of different things, including angiogenesis,” Dr Lin said.
“We used CD31 immunohistochemistry to look at angiogenesis, and, in the untreated mice, we didn’t see a lot of staining. But in the tumor-bearing mice, there was a lot more staining [P=0.0231]. And when we gave the mice tasquinimod, angiogenesis was significantly decreased [P<0.0001].”
The researchers also looked at different angiogenic factors. And they found that, compared to control-treated mice with MM, tumor-bearing mice that received tasquinimod had a significant decrease in serum levels of VEGF, FGF2, tissue factor, and endoglin.
The team is now assessing the effects of S100A9 and tasquinimod on megakaryocytes and platelets, 2 of the major cell populations that promote angiogenesis.
*Information in the abstract differs from that presented at the meeting.
PHILADELPHIA—A drug that has fallen short of expectations in clinical trials of prostate cancer may be effective for treating multiple myeloma (MM), according to research presented at the AACR Annual Meeting 2015.
The drug, tasquinimod, inhibits the function of S100A9, a pro-inflammatory protein that is elevated in MM, prostate cancer, and other malignancies.
Researchers found that tasquinimod can reduce tumor growth and improve survival in mouse models of MM. And these effects are associated with reduced angiogenesis in the bone marrow.
Cindy Lin, PhD, of the Wistar Institute in Philadelphia, Pennsylvania, and her colleagues presented these findings as abstract 1364.* The research was supported by Active Biotech and Ipsen, the companies developing tasquinimod.
Dr Lin noted that tasquinimod has already been tested in clinical trials of prostate cancer and initially appeared to be very effective. However, recent results from a phase 3 trial suggested the drug does not confer a favorable risk-benefit ratio for this population.
So Active Biotech and Ipsen decided to discontinue all trials of tasquinimod in prostate cancer. But the preclinical results observed in MM suggest tasquinimod may hold promise for treating these patients.
Activity in MM
Dr Lin said previous preclinical experiments revealed that myeloid-derived suppressor cells are involved in regulating MM progression, and these cells produce S100A9.
Tasquinimod is a quinoline-3-carboxamide derivative that binds to S100A9 and inhibits interaction with its receptors. So Dr Lin and her colleagues decided to investigate the antitumor effect of the drug in mouse models of MM.
The researchers initially tested tasquinimod in a syngeneic MM model, randomizing mice to treatment or control. Mice in the treatment group received tasquinimod at 30 mg/kg/day in their drinking water for 28 days.
Tasquinimod significantly improved survival in this model (P<0.005). All control mice died within 30 days of tumor inoculation, but about 40% of tasquinimod-treated mice were still alive more than 80 days out.
The researchers then tested tasquinimod in xenograft models of human MM. The drug significantly reduced tumor size in both H929 (P=0.0042) and RPMI-8226 models (P=0.0003). Treatment significantly improved survival in the H929 (P=0.0008) and RPMI-8226 models as well (P=0.0243).
Dr Lin said she and her colleagues did not see any side effects of treatment in any of the mice.
Investigating the mechanism
To determine if the antitumor effect of tasquinimod is, in fact, mediated through inhibition of S100A9, the researchers administered the drug to S100A9-knockout mice with MM. Tasquinimod did not improve survival in these mice, which suggests its anti-MM effects are mediated through S100A9 inhibition.
“To try and investigate some of the mechanisms of how survival is improved in tasquinimod-treated, tumor-bearing mice, we looked at a variety of different things, including angiogenesis,” Dr Lin said.
“We used CD31 immunohistochemistry to look at angiogenesis, and, in the untreated mice, we didn’t see a lot of staining. But in the tumor-bearing mice, there was a lot more staining [P=0.0231]. And when we gave the mice tasquinimod, angiogenesis was significantly decreased [P<0.0001].”
The researchers also looked at different angiogenic factors. And they found that, compared to control-treated mice with MM, tumor-bearing mice that received tasquinimod had a significant decrease in serum levels of VEGF, FGF2, tissue factor, and endoglin.
The team is now assessing the effects of S100A9 and tasquinimod on megakaryocytes and platelets, 2 of the major cell populations that promote angiogenesis.
*Information in the abstract differs from that presented at the meeting.
Using cervical length screening to predict preterm birth
One of the key indicators of a nation’s health is how well it can care for its young. Despite many advances in medical care and improvements in access to care, infant mortality remains a significant concern worldwide. According to the World Health Organization, the leading cause of death among children under age 5 is preterm birth complications. With an estimated 15 million babies born prematurely (prior to 37 weeks’ gestation) globally each year, it is vital for ob.gyns. to uncover ways to predict, diagnose early, and treat the causes of preterm birth.
While the challenges to infant health could be considered more of an issue in developing countries, here in the United States, the Centers for Disease Control and Prevention estimates that 1 in 9 babies is born prematurely. Preterm birth-related causes of death (i.e., breathing and feeding problems and disabilities) accounted for 35% of all infant deaths in 2010.
The World Health Organization (WHO) lists the United States as one of the top 10 countries with the greatest number of preterm births, despite the fact that we spend approximately 17.1% of our gross domestic product in total health care expenditures – the highest rate among our peer nations.
In the April 2014 edition of Master Class, we discussed one of the primary causes of preterm birth, bacterial infections, and specifically the need for ob.gyns. to rigorously screen patients for asymptomatic bacteriuria, which can lead to pyelonephritis. This month, we examine another biologic marker of preterm birth, cervical length.
Seminal studies of transvaginal sonography to measure cervical length during pregnancy and predict premature birth were published more than 2 decades ago. This work showed that a short cervix at 24 and 28 weeks’ gestation predicted preterm birth. Since then, clinical studies have demonstrated the utility of cervical length screening in women with prior preterm pregnancies. In the last decade, three large, randomized human trials have examined the usefulness of universal cervical length screening (Am. J. Obstet. Gynecol. 2012;207:101-6). However, the results of these trials have given practitioners a confusing picture of the predictability of this biologic marker.
Given the complexity of the “to screen or not to screen” issue, we have devoted this Master Class to a discussion on the role of cervical length screening and the prediction of preterm birth. Our guest author this month is Dr. Erika Werner, an assistant professor in ob.gyn (maternal-fetal medicine) in the department of obstetrics and gynecology at Brown University, in Providence, R.I., and an expert in the area of preterm birth.
Dr. Reece, who specializes in maternal-fetal medicine, is vice president for medical affairs at the University of Maryland, Baltimore, as well as the John Z. and Akiko K. Bowers Distinguished Professor and dean of the school of medicine. Dr. Reece said he had no relevant financial disclosures. He is the medical editor of this column. Contact him at [email protected].
One of the key indicators of a nation’s health is how well it can care for its young. Despite many advances in medical care and improvements in access to care, infant mortality remains a significant concern worldwide. According to the World Health Organization, the leading cause of death among children under age 5 is preterm birth complications. With an estimated 15 million babies born prematurely (prior to 37 weeks’ gestation) globally each year, it is vital for ob.gyns. to uncover ways to predict, diagnose early, and treat the causes of preterm birth.
While the challenges to infant health could be considered more of an issue in developing countries, here in the United States, the Centers for Disease Control and Prevention estimates that 1 in 9 babies is born prematurely. Preterm birth-related causes of death (i.e., breathing and feeding problems and disabilities) accounted for 35% of all infant deaths in 2010.
The World Health Organization (WHO) lists the United States as one of the top 10 countries with the greatest number of preterm births, despite the fact that we spend approximately 17.1% of our gross domestic product in total health care expenditures – the highest rate among our peer nations.
In the April 2014 edition of Master Class, we discussed one of the primary causes of preterm birth, bacterial infections, and specifically the need for ob.gyns. to rigorously screen patients for asymptomatic bacteriuria, which can lead to pyelonephritis. This month, we examine another biologic marker of preterm birth, cervical length.
Seminal studies of transvaginal sonography to measure cervical length during pregnancy and predict premature birth were published more than 2 decades ago. This work showed that a short cervix at 24 and 28 weeks’ gestation predicted preterm birth. Since then, clinical studies have demonstrated the utility of cervical length screening in women with prior preterm pregnancies. In the last decade, three large, randomized human trials have examined the usefulness of universal cervical length screening (Am. J. Obstet. Gynecol. 2012;207:101-6). However, the results of these trials have given practitioners a confusing picture of the predictability of this biologic marker.
Given the complexity of the “to screen or not to screen” issue, we have devoted this Master Class to a discussion on the role of cervical length screening and the prediction of preterm birth. Our guest author this month is Dr. Erika Werner, an assistant professor in ob.gyn (maternal-fetal medicine) in the department of obstetrics and gynecology at Brown University, in Providence, R.I., and an expert in the area of preterm birth.
Dr. Reece, who specializes in maternal-fetal medicine, is vice president for medical affairs at the University of Maryland, Baltimore, as well as the John Z. and Akiko K. Bowers Distinguished Professor and dean of the school of medicine. Dr. Reece said he had no relevant financial disclosures. He is the medical editor of this column. Contact him at [email protected].
One of the key indicators of a nation’s health is how well it can care for its young. Despite many advances in medical care and improvements in access to care, infant mortality remains a significant concern worldwide. According to the World Health Organization, the leading cause of death among children under age 5 is preterm birth complications. With an estimated 15 million babies born prematurely (prior to 37 weeks’ gestation) globally each year, it is vital for ob.gyns. to uncover ways to predict, diagnose early, and treat the causes of preterm birth.
While the challenges to infant health could be considered more of an issue in developing countries, here in the United States, the Centers for Disease Control and Prevention estimates that 1 in 9 babies is born prematurely. Preterm birth-related causes of death (i.e., breathing and feeding problems and disabilities) accounted for 35% of all infant deaths in 2010.
The World Health Organization (WHO) lists the United States as one of the top 10 countries with the greatest number of preterm births, despite the fact that we spend approximately 17.1% of our gross domestic product in total health care expenditures – the highest rate among our peer nations.
In the April 2014 edition of Master Class, we discussed one of the primary causes of preterm birth, bacterial infections, and specifically the need for ob.gyns. to rigorously screen patients for asymptomatic bacteriuria, which can lead to pyelonephritis. This month, we examine another biologic marker of preterm birth, cervical length.
Seminal studies of transvaginal sonography to measure cervical length during pregnancy and predict premature birth were published more than 2 decades ago. This work showed that a short cervix at 24 and 28 weeks’ gestation predicted preterm birth. Since then, clinical studies have demonstrated the utility of cervical length screening in women with prior preterm pregnancies. In the last decade, three large, randomized human trials have examined the usefulness of universal cervical length screening (Am. J. Obstet. Gynecol. 2012;207:101-6). However, the results of these trials have given practitioners a confusing picture of the predictability of this biologic marker.
Given the complexity of the “to screen or not to screen” issue, we have devoted this Master Class to a discussion on the role of cervical length screening and the prediction of preterm birth. Our guest author this month is Dr. Erika Werner, an assistant professor in ob.gyn (maternal-fetal medicine) in the department of obstetrics and gynecology at Brown University, in Providence, R.I., and an expert in the area of preterm birth.
Dr. Reece, who specializes in maternal-fetal medicine, is vice president for medical affairs at the University of Maryland, Baltimore, as well as the John Z. and Akiko K. Bowers Distinguished Professor and dean of the school of medicine. Dr. Reece said he had no relevant financial disclosures. He is the medical editor of this column. Contact him at [email protected].
The benefits, costs of universal cervical length screening
Rates of preterm birth in the United States have been falling since 2006, but the rates of early preterm birth in singletons (those under 34 weeks’ gestation), specifically, have not trended downward as dramatically as have late preterm birth in singletons (34-36 weeks). According to 2015 data from the National Vital Statistics Reports, the rate of early preterm births is still 3.4% in all pregnancies and 2.7% among singletons.
While the number of neonates born before 37 weeks of gestation remains high – approximately 11% in 2013 – and signifies a continuing public health problem, the rate of early preterm birth is particularly concerning because early preterm birth is more significantly associated with neonatal mortality, long-term morbidity and extended neonatal intensive care unit stays, all leading to increased health care expenditures.
Finding predictors for preterm birth that are stronger than traditional clinical factors has long been a goal of ob.gyns. because the vast majority of all spontaneous preterm births occur to women without known risk factors (i.e., multiple gestations or prior preterm birth).
Cervical length in the midtrimester is now a well-verified predictor of preterm birth, for both low- and high-risk women. Furthermore, vaginal progesterone has been shown to be a safe and beneficial intervention for women with no known risk factors who are diagnosed with a shortened cervical length (< 2 cm), and cervical cerclage has been suggested to reduce the risk of preterm birth for women with a history of prior preterm birth who also have a shortened cervical length.
Some are now advocating universal cervical length screening for women with singleton gestations, but before universal screening is mandated, the downstream effect of such a change in practice must be considered.
Backdrop to screening
Cervical length measurement was first investigated more than 25 years ago as a possible predictor of preterm birth. In 1996, a prospective multicenter study of almost 3,000 women with singleton pregnancies showed that the risk of preterm delivery is inversely and directly related to the length of the cervix, as measured with vaginal ultrasonography (N. Engl. J. Med. 1996;334:567-72).
In fact, at 24 weeks’ gestation, every 1 mm of additional cervical length equates to a significant decrease in preterm birth risk (odds ratio, 0.91). Several other studies, in addition to the landmark 1996 study, have similarly demonstrated this inverse relationship between preterm birth risk and cervical length between 18 and 24 weeks’ gestation.
However, the use of cervical measurement did not achieve widespread use until more than a decade later, when researchers began to identify interventions that could prolong pregnancy if a short cervix was diagnosed in the second trimester.
For example, Dr. E.B. Fonseca’s study of almost 25,000 asymptomatic pregnant women, demonstrated that daily vaginal progesterone reduced the risk of spontaneous delivery before 34 weeks by approximately 44% in women identified with a cervical length of 1.5 cm or less (N. Engl. J. Med. 2007;357:462-9). The vast majority of the women in this study had singleton pregnancies.
Shortly thereafter, Dr. S.S. Hassan and her colleagues completed a similar trial in women with singleton gestations and transvaginal cervical lengths between 1.0 and 2.0 cm at 20-23 weeks’ gestation. In this trial, nightly progesterone gel (with 90 mg progesterone per application) was associated with a 45% reduction in preterm birth before 33 weeks and a 38% reduction in preterm birth before 35 weeks (Ultrasound. Obstet. Gynecol. 2011;38:18-31).
A meta-analysis led by Dr. Roberto Romero, which included the Fonseca and Hassan trials, looked specifically at 775 women with a midtrimester cervical length of 2.5 cm or less. Women with a singleton gestation who had no history of preterm birth had a 40% reduction in the rate of early preterm birth when they were treated with vaginal progesterone (Am. J. Obstet. Gynecol. 2012;206:124-e1-19).
The benefits of identifying a short cervix likely extend to women with a history of prior preterm birth. A patient-level meta-analysis published in 2011 demonstrated that cervical cerclage placement was associated with a significant reduction in preterm birth before 35 weeks’ gestation in women with singleton gestations, previous spontaneous preterm birth, and cervical length less than 2.5 cm before 24 weeks’ gestation (Obstet. Gynecol. 2011;117:663-71).
The possible benefits of diagnosing and intervening for a shortened cervix have tipped many experts and clinicians toward the practice of universal cervical length screening of all singleton pregnancies. Research has shown that we can accurately obtain a cervical-length measurement before 24 weeks, and that we have effective and safe interventions for cases of short cervix: cerclage in women with a history of preterm birth who are already receiving progesterone, and vaginal progesterone in women without such a history.
Screening certainties and doubts
In 2011, my colleagues and I compared the cost effectiveness of two approaches to preterm birth prevention in low-risk pregnancies: no screening versus a single transvaginal ultrasound cervical-length measurement in all asymptomatic, low-risk singleton pregnant individuals between 18 and 24 weeks’ gestation.
In our model, women identified as having a cervical length less than 1.5 cm would be offered vaginal progesterone. Based on published data, we assumed there would be a 92% adherence rate, and a 45% reduction in deliveries before 34 weeks with progesterone treatment.
We found that in low-risk pregnancies, universal transvaginal cervical-length ultrasound screening and progesterone intervention would be cost effective and in many cases cost saving. We estimated that screening would prevent 248 early preterm births – as well as 22 neonatal deaths or neonates with long-term neurologic deficits – per 100,000 deliveries.
Our sensitivity analyses showed that screening remained cost saving under a range of clinical scenarios, including varied preterm birth rates and predictive values of a shortened cervix. Screening was not cost saving, but remained cost effective, when the expense of a transvaginal ultrasound scan exceeds $187 or when vaginal progesterone is assumed to reduce the risk of early preterm delivery by less than 20% (Ultrasound Obstet. Gynecol. 2011;38;32-37).
Neither the American College of Obstetricians and Gynecologists nor the Society for Maternal-Fetal Medicine support mandated universal transvaginal ultrasound cervical length screening. Both organizations state, however, that the approach may be considered in women with singleton gestations without prior spontaneous preterm birth.
Interestingly, Thomas Jefferson University in Philadelphia, which uses a universal screening program for singleton gestations without prior preterm birth, has recently published data that complicate the growing trend toward universal cervical length screening.
The Philadelphia clinicians followed a strategy whereby women with a transvaginal cervical length of 2 cm or less were prescribed vaginal progesterone (90 mg vaginal progesterone gel, or 200 mg micronized progesterone gel capsules). Those with a cervical length between approximately 2 cm and 2.5 cm were asked to return for a follow-up cervical length measurement before 24 weeks’ gestation.
What they found in this cohort was surprising: a rate of short cervix that is significantly lower than what previous research has shown.
Among those screened, 0.8% of women had a cervical length of 2 cm or less on an initial transvaginal ultrasonogram. Previously, a prevalence of 1%-2% for an even shorter cervical length (less than 1.5 cm) was fairly consistent in the literature.
As Dr. Kelly M. Orzechowski and her colleagues point out, the low incidence of short cervix “raises questions regarding whether universal transvaginal ultrasonogram cervical length screening in low-risk asymptomatic women is beneficial” (Obstet. Gynecol. 2014;124:520-5).
In our 2011 cost-effectiveness analysis, we found that screening was no longer a cost-saving practice when the incidence of cervical length less than 1.5 cm falls below 0.8%. Screening remained cost effective, however.
Recently, we found that if the Philadelphia protocol is followed and the U.S. population has an incidence of shortened cervix similar to that described by Dr. Orzechowski and her colleagues, universal cervical length screening in low-risk singleton pregnancies is cost effective but not cost saving. Furthermore, we found several additional plausible situations in this unpublished analysis in which universal screening ceased to be cost effective.
Thus, before we move to a strategy of mandated universal screening, we need better population-based estimates of the incidence of short cervix in a truly low-risk population.
We also must consider the future costs of progesterone. It is possible that costs may increase significantly if vaginal progesterone wins approval from the Food and Drug Administration for this indication.
Finally, if universal cervical length screening is to become the standard of care, we need policies in place to prevent misuse of the screening technology that would inevitably drive up costs without improving outcomes. For example, we must ensure that one cervical length measurement does not transition into serial cervical length measurements over the course of pregnancy, since measurement after 24 weeks has limited clinical utility. Similarly, progesterone use for a cervical length less than or equal to 2.0 cm cannot progress to progesterone for anyone approaching 2.0 cm (i.e. 2.5 cm or even 3 cm) as there is no evidence to suggest a benefit for women with longer cervixes.
Over time, it would be beneficial to have additional data on how best to manage patients who have a cervical length of 2 cm-2.5 cm before 24 weeks’ gestation. Many of us ask these women to return for a follow-up measurement and some may prescribe progesterone. However, we lack evidence for either approach; while a cervical length measurement less than 2.5 cm is clearly associated with an increased risk of preterm birth, the benefit of treatment has been demonstrated only with a cervical length of 2 cm or less.
Today and the future
For women with a history of preterm birth, cervical length screening is now routine. For low-risk pregnant women – those without a history of previous spontaneous preterm delivery – various approaches are currently taken. Most physicians recommend assessing the cervical length transabdominally at the time of the 18-20-week ultrasound, and proceeding to transvaginal ultrasonography if the cervical length is less than 3 cm or 3.5 cm.
To reliably image the cervix with transabdominal ultrasound, it should be performed with a full bladder and with the understanding that the cervix appears longer (6 mm longer, on average) when the bladder is full (Aust. N. Z. J. Obstet. Gynaecol. 2014;54:250-55).
Transvaginal ultrasound has been widely recognized as a sensitive and reproducible method for detecting shortened cervical length. Overall, this tool has several advantages over the transabdominal approach. However, the lack of universal access to transvaginal ultrasound and to consistently reliable cervical length measurements have been valid concerns of those who oppose universal transvaginal ultrasound cervical length screening.
Such concerns likely will lessen over time as transvaginal ultrasound continues to become more pervasive. Several years ago, the Perinatal Quality Foundation set standards for measuring the cervix and launched the Cervical Length Education and Review (CLEAR) program. When sonographers and physicians obtain training and credentialing, there appears to be only a 5%-10% intraobserver variability in cervical length measurement. (The PQF’s initial focus in 2005 was the Nuchal Translucency Quality Review program.)
Increasingly, I believe, transvaginal ultrasound cervical length measurement will be utilized to identify women at high risk for early preterm birth so that low-risk women can receive progesterone and high-risk women (those with a history of preterm birth) can be considered as candidates for cerclage placement. In the process, the quality of clinical care as well as the quality of our research data will improve. Whether and when such screening will become universal, however, is still uncertain.
Dr. Werner reported that she has no financial disclosures relevant to this Master Class.
Rates of preterm birth in the United States have been falling since 2006, but the rates of early preterm birth in singletons (those under 34 weeks’ gestation), specifically, have not trended downward as dramatically as have late preterm birth in singletons (34-36 weeks). According to 2015 data from the National Vital Statistics Reports, the rate of early preterm births is still 3.4% in all pregnancies and 2.7% among singletons.
While the number of neonates born before 37 weeks of gestation remains high – approximately 11% in 2013 – and signifies a continuing public health problem, the rate of early preterm birth is particularly concerning because early preterm birth is more significantly associated with neonatal mortality, long-term morbidity and extended neonatal intensive care unit stays, all leading to increased health care expenditures.
Finding predictors for preterm birth that are stronger than traditional clinical factors has long been a goal of ob.gyns. because the vast majority of all spontaneous preterm births occur to women without known risk factors (i.e., multiple gestations or prior preterm birth).
Cervical length in the midtrimester is now a well-verified predictor of preterm birth, for both low- and high-risk women. Furthermore, vaginal progesterone has been shown to be a safe and beneficial intervention for women with no known risk factors who are diagnosed with a shortened cervical length (< 2 cm), and cervical cerclage has been suggested to reduce the risk of preterm birth for women with a history of prior preterm birth who also have a shortened cervical length.
Some are now advocating universal cervical length screening for women with singleton gestations, but before universal screening is mandated, the downstream effect of such a change in practice must be considered.
Backdrop to screening
Cervical length measurement was first investigated more than 25 years ago as a possible predictor of preterm birth. In 1996, a prospective multicenter study of almost 3,000 women with singleton pregnancies showed that the risk of preterm delivery is inversely and directly related to the length of the cervix, as measured with vaginal ultrasonography (N. Engl. J. Med. 1996;334:567-72).
In fact, at 24 weeks’ gestation, every 1 mm of additional cervical length equates to a significant decrease in preterm birth risk (odds ratio, 0.91). Several other studies, in addition to the landmark 1996 study, have similarly demonstrated this inverse relationship between preterm birth risk and cervical length between 18 and 24 weeks’ gestation.
However, the use of cervical measurement did not achieve widespread use until more than a decade later, when researchers began to identify interventions that could prolong pregnancy if a short cervix was diagnosed in the second trimester.
For example, Dr. E.B. Fonseca’s study of almost 25,000 asymptomatic pregnant women, demonstrated that daily vaginal progesterone reduced the risk of spontaneous delivery before 34 weeks by approximately 44% in women identified with a cervical length of 1.5 cm or less (N. Engl. J. Med. 2007;357:462-9). The vast majority of the women in this study had singleton pregnancies.
Shortly thereafter, Dr. S.S. Hassan and her colleagues completed a similar trial in women with singleton gestations and transvaginal cervical lengths between 1.0 and 2.0 cm at 20-23 weeks’ gestation. In this trial, nightly progesterone gel (with 90 mg progesterone per application) was associated with a 45% reduction in preterm birth before 33 weeks and a 38% reduction in preterm birth before 35 weeks (Ultrasound. Obstet. Gynecol. 2011;38:18-31).
A meta-analysis led by Dr. Roberto Romero, which included the Fonseca and Hassan trials, looked specifically at 775 women with a midtrimester cervical length of 2.5 cm or less. Women with a singleton gestation who had no history of preterm birth had a 40% reduction in the rate of early preterm birth when they were treated with vaginal progesterone (Am. J. Obstet. Gynecol. 2012;206:124-e1-19).
The benefits of identifying a short cervix likely extend to women with a history of prior preterm birth. A patient-level meta-analysis published in 2011 demonstrated that cervical cerclage placement was associated with a significant reduction in preterm birth before 35 weeks’ gestation in women with singleton gestations, previous spontaneous preterm birth, and cervical length less than 2.5 cm before 24 weeks’ gestation (Obstet. Gynecol. 2011;117:663-71).
The possible benefits of diagnosing and intervening for a shortened cervix have tipped many experts and clinicians toward the practice of universal cervical length screening of all singleton pregnancies. Research has shown that we can accurately obtain a cervical-length measurement before 24 weeks, and that we have effective and safe interventions for cases of short cervix: cerclage in women with a history of preterm birth who are already receiving progesterone, and vaginal progesterone in women without such a history.
Screening certainties and doubts
In 2011, my colleagues and I compared the cost effectiveness of two approaches to preterm birth prevention in low-risk pregnancies: no screening versus a single transvaginal ultrasound cervical-length measurement in all asymptomatic, low-risk singleton pregnant individuals between 18 and 24 weeks’ gestation.
In our model, women identified as having a cervical length less than 1.5 cm would be offered vaginal progesterone. Based on published data, we assumed there would be a 92% adherence rate, and a 45% reduction in deliveries before 34 weeks with progesterone treatment.
We found that in low-risk pregnancies, universal transvaginal cervical-length ultrasound screening and progesterone intervention would be cost effective and in many cases cost saving. We estimated that screening would prevent 248 early preterm births – as well as 22 neonatal deaths or neonates with long-term neurologic deficits – per 100,000 deliveries.
Our sensitivity analyses showed that screening remained cost saving under a range of clinical scenarios, including varied preterm birth rates and predictive values of a shortened cervix. Screening was not cost saving, but remained cost effective, when the expense of a transvaginal ultrasound scan exceeds $187 or when vaginal progesterone is assumed to reduce the risk of early preterm delivery by less than 20% (Ultrasound Obstet. Gynecol. 2011;38;32-37).
Neither the American College of Obstetricians and Gynecologists nor the Society for Maternal-Fetal Medicine support mandated universal transvaginal ultrasound cervical length screening. Both organizations state, however, that the approach may be considered in women with singleton gestations without prior spontaneous preterm birth.
Interestingly, Thomas Jefferson University in Philadelphia, which uses a universal screening program for singleton gestations without prior preterm birth, has recently published data that complicate the growing trend toward universal cervical length screening.
The Philadelphia clinicians followed a strategy whereby women with a transvaginal cervical length of 2 cm or less were prescribed vaginal progesterone (90 mg vaginal progesterone gel, or 200 mg micronized progesterone gel capsules). Those with a cervical length between approximately 2 cm and 2.5 cm were asked to return for a follow-up cervical length measurement before 24 weeks’ gestation.
What they found in this cohort was surprising: a rate of short cervix that is significantly lower than what previous research has shown.
Among those screened, 0.8% of women had a cervical length of 2 cm or less on an initial transvaginal ultrasonogram. Previously, a prevalence of 1%-2% for an even shorter cervical length (less than 1.5 cm) was fairly consistent in the literature.
As Dr. Kelly M. Orzechowski and her colleagues point out, the low incidence of short cervix “raises questions regarding whether universal transvaginal ultrasonogram cervical length screening in low-risk asymptomatic women is beneficial” (Obstet. Gynecol. 2014;124:520-5).
In our 2011 cost-effectiveness analysis, we found that screening was no longer a cost-saving practice when the incidence of cervical length less than 1.5 cm falls below 0.8%. Screening remained cost effective, however.
Recently, we found that if the Philadelphia protocol is followed and the U.S. population has an incidence of shortened cervix similar to that described by Dr. Orzechowski and her colleagues, universal cervical length screening in low-risk singleton pregnancies is cost effective but not cost saving. Furthermore, we found several additional plausible situations in this unpublished analysis in which universal screening ceased to be cost effective.
Thus, before we move to a strategy of mandated universal screening, we need better population-based estimates of the incidence of short cervix in a truly low-risk population.
We also must consider the future costs of progesterone. It is possible that costs may increase significantly if vaginal progesterone wins approval from the Food and Drug Administration for this indication.
Finally, if universal cervical length screening is to become the standard of care, we need policies in place to prevent misuse of the screening technology that would inevitably drive up costs without improving outcomes. For example, we must ensure that one cervical length measurement does not transition into serial cervical length measurements over the course of pregnancy, since measurement after 24 weeks has limited clinical utility. Similarly, progesterone use for a cervical length less than or equal to 2.0 cm cannot progress to progesterone for anyone approaching 2.0 cm (i.e. 2.5 cm or even 3 cm) as there is no evidence to suggest a benefit for women with longer cervixes.
Over time, it would be beneficial to have additional data on how best to manage patients who have a cervical length of 2 cm-2.5 cm before 24 weeks’ gestation. Many of us ask these women to return for a follow-up measurement and some may prescribe progesterone. However, we lack evidence for either approach; while a cervical length measurement less than 2.5 cm is clearly associated with an increased risk of preterm birth, the benefit of treatment has been demonstrated only with a cervical length of 2 cm or less.
Today and the future
For women with a history of preterm birth, cervical length screening is now routine. For low-risk pregnant women – those without a history of previous spontaneous preterm delivery – various approaches are currently taken. Most physicians recommend assessing the cervical length transabdominally at the time of the 18-20-week ultrasound, and proceeding to transvaginal ultrasonography if the cervical length is less than 3 cm or 3.5 cm.
To reliably image the cervix with transabdominal ultrasound, it should be performed with a full bladder and with the understanding that the cervix appears longer (6 mm longer, on average) when the bladder is full (Aust. N. Z. J. Obstet. Gynaecol. 2014;54:250-55).
Transvaginal ultrasound has been widely recognized as a sensitive and reproducible method for detecting shortened cervical length. Overall, this tool has several advantages over the transabdominal approach. However, the lack of universal access to transvaginal ultrasound and to consistently reliable cervical length measurements have been valid concerns of those who oppose universal transvaginal ultrasound cervical length screening.
Such concerns likely will lessen over time as transvaginal ultrasound continues to become more pervasive. Several years ago, the Perinatal Quality Foundation set standards for measuring the cervix and launched the Cervical Length Education and Review (CLEAR) program. When sonographers and physicians obtain training and credentialing, there appears to be only a 5%-10% intraobserver variability in cervical length measurement. (The PQF’s initial focus in 2005 was the Nuchal Translucency Quality Review program.)
Increasingly, I believe, transvaginal ultrasound cervical length measurement will be utilized to identify women at high risk for early preterm birth so that low-risk women can receive progesterone and high-risk women (those with a history of preterm birth) can be considered as candidates for cerclage placement. In the process, the quality of clinical care as well as the quality of our research data will improve. Whether and when such screening will become universal, however, is still uncertain.
Dr. Werner reported that she has no financial disclosures relevant to this Master Class.
Rates of preterm birth in the United States have been falling since 2006, but the rates of early preterm birth in singletons (those under 34 weeks’ gestation), specifically, have not trended downward as dramatically as have late preterm birth in singletons (34-36 weeks). According to 2015 data from the National Vital Statistics Reports, the rate of early preterm births is still 3.4% in all pregnancies and 2.7% among singletons.
While the number of neonates born before 37 weeks of gestation remains high – approximately 11% in 2013 – and signifies a continuing public health problem, the rate of early preterm birth is particularly concerning because early preterm birth is more significantly associated with neonatal mortality, long-term morbidity and extended neonatal intensive care unit stays, all leading to increased health care expenditures.
Finding predictors for preterm birth that are stronger than traditional clinical factors has long been a goal of ob.gyns. because the vast majority of all spontaneous preterm births occur to women without known risk factors (i.e., multiple gestations or prior preterm birth).
Cervical length in the midtrimester is now a well-verified predictor of preterm birth, for both low- and high-risk women. Furthermore, vaginal progesterone has been shown to be a safe and beneficial intervention for women with no known risk factors who are diagnosed with a shortened cervical length (< 2 cm), and cervical cerclage has been suggested to reduce the risk of preterm birth for women with a history of prior preterm birth who also have a shortened cervical length.
Some are now advocating universal cervical length screening for women with singleton gestations, but before universal screening is mandated, the downstream effect of such a change in practice must be considered.
Backdrop to screening
Cervical length measurement was first investigated more than 25 years ago as a possible predictor of preterm birth. In 1996, a prospective multicenter study of almost 3,000 women with singleton pregnancies showed that the risk of preterm delivery is inversely and directly related to the length of the cervix, as measured with vaginal ultrasonography (N. Engl. J. Med. 1996;334:567-72).
In fact, at 24 weeks’ gestation, every 1 mm of additional cervical length equates to a significant decrease in preterm birth risk (odds ratio, 0.91). Several other studies, in addition to the landmark 1996 study, have similarly demonstrated this inverse relationship between preterm birth risk and cervical length between 18 and 24 weeks’ gestation.
However, the use of cervical measurement did not achieve widespread use until more than a decade later, when researchers began to identify interventions that could prolong pregnancy if a short cervix was diagnosed in the second trimester.
For example, Dr. E.B. Fonseca’s study of almost 25,000 asymptomatic pregnant women, demonstrated that daily vaginal progesterone reduced the risk of spontaneous delivery before 34 weeks by approximately 44% in women identified with a cervical length of 1.5 cm or less (N. Engl. J. Med. 2007;357:462-9). The vast majority of the women in this study had singleton pregnancies.
Shortly thereafter, Dr. S.S. Hassan and her colleagues completed a similar trial in women with singleton gestations and transvaginal cervical lengths between 1.0 and 2.0 cm at 20-23 weeks’ gestation. In this trial, nightly progesterone gel (with 90 mg progesterone per application) was associated with a 45% reduction in preterm birth before 33 weeks and a 38% reduction in preterm birth before 35 weeks (Ultrasound. Obstet. Gynecol. 2011;38:18-31).
A meta-analysis led by Dr. Roberto Romero, which included the Fonseca and Hassan trials, looked specifically at 775 women with a midtrimester cervical length of 2.5 cm or less. Women with a singleton gestation who had no history of preterm birth had a 40% reduction in the rate of early preterm birth when they were treated with vaginal progesterone (Am. J. Obstet. Gynecol. 2012;206:124-e1-19).
The benefits of identifying a short cervix likely extend to women with a history of prior preterm birth. A patient-level meta-analysis published in 2011 demonstrated that cervical cerclage placement was associated with a significant reduction in preterm birth before 35 weeks’ gestation in women with singleton gestations, previous spontaneous preterm birth, and cervical length less than 2.5 cm before 24 weeks’ gestation (Obstet. Gynecol. 2011;117:663-71).
The possible benefits of diagnosing and intervening for a shortened cervix have tipped many experts and clinicians toward the practice of universal cervical length screening of all singleton pregnancies. Research has shown that we can accurately obtain a cervical-length measurement before 24 weeks, and that we have effective and safe interventions for cases of short cervix: cerclage in women with a history of preterm birth who are already receiving progesterone, and vaginal progesterone in women without such a history.
Screening certainties and doubts
In 2011, my colleagues and I compared the cost effectiveness of two approaches to preterm birth prevention in low-risk pregnancies: no screening versus a single transvaginal ultrasound cervical-length measurement in all asymptomatic, low-risk singleton pregnant individuals between 18 and 24 weeks’ gestation.
In our model, women identified as having a cervical length less than 1.5 cm would be offered vaginal progesterone. Based on published data, we assumed there would be a 92% adherence rate, and a 45% reduction in deliveries before 34 weeks with progesterone treatment.
We found that in low-risk pregnancies, universal transvaginal cervical-length ultrasound screening and progesterone intervention would be cost effective and in many cases cost saving. We estimated that screening would prevent 248 early preterm births – as well as 22 neonatal deaths or neonates with long-term neurologic deficits – per 100,000 deliveries.
Our sensitivity analyses showed that screening remained cost saving under a range of clinical scenarios, including varied preterm birth rates and predictive values of a shortened cervix. Screening was not cost saving, but remained cost effective, when the expense of a transvaginal ultrasound scan exceeds $187 or when vaginal progesterone is assumed to reduce the risk of early preterm delivery by less than 20% (Ultrasound Obstet. Gynecol. 2011;38;32-37).
Neither the American College of Obstetricians and Gynecologists nor the Society for Maternal-Fetal Medicine support mandated universal transvaginal ultrasound cervical length screening. Both organizations state, however, that the approach may be considered in women with singleton gestations without prior spontaneous preterm birth.
Interestingly, Thomas Jefferson University in Philadelphia, which uses a universal screening program for singleton gestations without prior preterm birth, has recently published data that complicate the growing trend toward universal cervical length screening.
The Philadelphia clinicians followed a strategy whereby women with a transvaginal cervical length of 2 cm or less were prescribed vaginal progesterone (90 mg vaginal progesterone gel, or 200 mg micronized progesterone gel capsules). Those with a cervical length between approximately 2 cm and 2.5 cm were asked to return for a follow-up cervical length measurement before 24 weeks’ gestation.
What they found in this cohort was surprising: a rate of short cervix that is significantly lower than what previous research has shown.
Among those screened, 0.8% of women had a cervical length of 2 cm or less on an initial transvaginal ultrasonogram. Previously, a prevalence of 1%-2% for an even shorter cervical length (less than 1.5 cm) was fairly consistent in the literature.
As Dr. Kelly M. Orzechowski and her colleagues point out, the low incidence of short cervix “raises questions regarding whether universal transvaginal ultrasonogram cervical length screening in low-risk asymptomatic women is beneficial” (Obstet. Gynecol. 2014;124:520-5).
In our 2011 cost-effectiveness analysis, we found that screening was no longer a cost-saving practice when the incidence of cervical length less than 1.5 cm falls below 0.8%. Screening remained cost effective, however.
Recently, we found that if the Philadelphia protocol is followed and the U.S. population has an incidence of shortened cervix similar to that described by Dr. Orzechowski and her colleagues, universal cervical length screening in low-risk singleton pregnancies is cost effective but not cost saving. Furthermore, we found several additional plausible situations in this unpublished analysis in which universal screening ceased to be cost effective.
Thus, before we move to a strategy of mandated universal screening, we need better population-based estimates of the incidence of short cervix in a truly low-risk population.
We also must consider the future costs of progesterone. It is possible that costs may increase significantly if vaginal progesterone wins approval from the Food and Drug Administration for this indication.
Finally, if universal cervical length screening is to become the standard of care, we need policies in place to prevent misuse of the screening technology that would inevitably drive up costs without improving outcomes. For example, we must ensure that one cervical length measurement does not transition into serial cervical length measurements over the course of pregnancy, since measurement after 24 weeks has limited clinical utility. Similarly, progesterone use for a cervical length less than or equal to 2.0 cm cannot progress to progesterone for anyone approaching 2.0 cm (i.e. 2.5 cm or even 3 cm) as there is no evidence to suggest a benefit for women with longer cervixes.
Over time, it would be beneficial to have additional data on how best to manage patients who have a cervical length of 2 cm-2.5 cm before 24 weeks’ gestation. Many of us ask these women to return for a follow-up measurement and some may prescribe progesterone. However, we lack evidence for either approach; while a cervical length measurement less than 2.5 cm is clearly associated with an increased risk of preterm birth, the benefit of treatment has been demonstrated only with a cervical length of 2 cm or less.
Today and the future
For women with a history of preterm birth, cervical length screening is now routine. For low-risk pregnant women – those without a history of previous spontaneous preterm delivery – various approaches are currently taken. Most physicians recommend assessing the cervical length transabdominally at the time of the 18-20-week ultrasound, and proceeding to transvaginal ultrasonography if the cervical length is less than 3 cm or 3.5 cm.
To reliably image the cervix with transabdominal ultrasound, it should be performed with a full bladder and with the understanding that the cervix appears longer (6 mm longer, on average) when the bladder is full (Aust. N. Z. J. Obstet. Gynaecol. 2014;54:250-55).
Transvaginal ultrasound has been widely recognized as a sensitive and reproducible method for detecting shortened cervical length. Overall, this tool has several advantages over the transabdominal approach. However, the lack of universal access to transvaginal ultrasound and to consistently reliable cervical length measurements have been valid concerns of those who oppose universal transvaginal ultrasound cervical length screening.
Such concerns likely will lessen over time as transvaginal ultrasound continues to become more pervasive. Several years ago, the Perinatal Quality Foundation set standards for measuring the cervix and launched the Cervical Length Education and Review (CLEAR) program. When sonographers and physicians obtain training and credentialing, there appears to be only a 5%-10% intraobserver variability in cervical length measurement. (The PQF’s initial focus in 2005 was the Nuchal Translucency Quality Review program.)
Increasingly, I believe, transvaginal ultrasound cervical length measurement will be utilized to identify women at high risk for early preterm birth so that low-risk women can receive progesterone and high-risk women (those with a history of preterm birth) can be considered as candidates for cerclage placement. In the process, the quality of clinical care as well as the quality of our research data will improve. Whether and when such screening will become universal, however, is still uncertain.
Dr. Werner reported that she has no financial disclosures relevant to this Master Class.
How to assess and treat birth-related depression in new fathers
Only recently has paternal postpartum depression (PPD) received much attention. Research has shown that maternal PPD is associated with negative outcomes in the child’s cognitive development and social and marital problems for the parents. Likewise, depressed fathers are less likely to play outside with their child and more likely to put the child to bed awake.1
Recent studies reported that 10.4% of men experienced depression within 12 month of delivery.1 Edmondson et al2 estimate the prevalence of paternal PPD to be 8% between birth and 3 months, 26% from 3 to 6 months, and 9% from 6 to 12 months.
Risk factors
Risk factors for paternal PPD have not been studied extensively. Some studies have shown that immaturity, lack of social support, first or unplanned pregnancies, marital relationship problems, and unemployment were the most common risk factors for depression in men postnatally.3 A history of depression and other psychiatric disorders also increases risk.4 Psychosocial factors, such as quality of the spousal relationship, parenting distress, and perceived parenting efficacy, contribute to paternal depression.
Similarly, depressed postpartum fathers experience higher levels of parenting distress and a lower sense of parenting efficacy.5 Interestingly, negative life events were associated with increased risk for depression in mothers, but not fathers.3
Clinical presentation
Paternal PPD symptoms appear within 12 months after the birth of the child and last for at least 2 weeks. Signs and symptoms of depression in men might not resemble those seen in postpartum women. Men tend to show aggression, increased or easy irritability, and agitation, and might not seek help for emotional issues as readily as women do. Typical symptoms of depression often are present, such as sleep disturbance or changes in sleep patterns, difficulty concentrating, memory problems, and feelings of worthlessness, hopelessness, inadequacy, and excess guilt with suicidal ideation.6
Making the diagnosis
Maternal PPD commonly is evaluated using the Edinburgh Postnatal Depression Scale- Partner (EDPS-P) or Postpartum Depression Screening Scale. However, studies are lacking to determine which diagnostic modality is most accurate for diagnosing paternal PPD.
A paternal PPD screening tool could include the EDPS-P administered to mothers. Edmonson et al2 determined an EDPS-P score of >10 was the optimal cut-off point for screening for paternal depression, with a sensitivity of 89.5% and a specificity of 78.2%, compared with a structured clinical interview. Fisher et al4 determined that the EDPS-P report was a reliable method for detecting paternal PPD compared with validated depression scales completed by fathers. Madsen et al5 determined the Gotland Male Depression Scale, which detects typical male depressive symptoms, also was effective in recognizing paternal PPD at 6 weeks postpartum.7
Treatment of paternal PPD
Specific treatment for paternal PPD has not been studied extensively. Psychotherapy targeted at interpersonal family relationships and parenting is indicated for mild depression, whereas a combination of psychotherapy and pharmacotherapy is recommended for moderate or severe depression.
Depending on specific patient factors, pharmacotherapy options include selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors, tricyclic antidepressants, and atypical antipsychotics.8 SSRIs often are used because of their efficacy and relative lack of serious side effects, as demonstrated in numerous trials.2 Recovery is more likely with combination therapy than monotherapy.9 Fathers with psychosis or suicidal ideation should be referred for inpatient treatment.
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Paulson JF, Dauber S, Leiferman JA. Individual and combined effects of postpartum depression in mothers and fathers on parenting behavior. Pediatrics. 2006;118(2):659-668.
2. Edmondson OJ, Psychogiou L, Vlachos H, et al. Depression in fathers in the postnatal period: assessment of the Edinburgh Postnatal Depression Scale as a screening measure. J Affect Disord. 2010;125(1-3):365-368.
3. Schumacher M, Zubaran C, White G. Bringing birth-related paternal depression to the fore. Women Birth. 2008;21(2):65-70.
4. Fisher SD, Kopelman R, O’Hara MW. Partner report of paternal depression using the Edinburgh Postnatal Depression Scale-Partner. Arch Womens Ment Health. 2012;15(4):283-288.
5. Madsen SA, Juhl T. Paternal depression in the postnatal period assessed with traditional and male depression scales. Journal of Men’s Health and Gender. 2007;4(1):26-31.
6. Escribà-Agüir V, Artazcoz L. Gender differences in postpartum depression: a longitudinal cohort study. J Epidemiol Community Health. 2011;65(4):320-326.
7. Cuijpers P, van Straten A, Warmerdam L, et al. Psychotherapy versus the combination of psychotherapy and pharmacotherapy in the treatment of depression: a meta-analysis. Depress Anxiety. 2009;26(3):279-288.
8. Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 12 new-generation anti-depressants: a multiple-treatments meta-analysis. Lancet. 2009;373(9665):746-758.
9. Demontigny F, Girard ME, Lacharité C, et al. Psychosocial factors associated with paternal postnatal depression. J Affect Disord. 2013;15(150):44-49.
Only recently has paternal postpartum depression (PPD) received much attention. Research has shown that maternal PPD is associated with negative outcomes in the child’s cognitive development and social and marital problems for the parents. Likewise, depressed fathers are less likely to play outside with their child and more likely to put the child to bed awake.1
Recent studies reported that 10.4% of men experienced depression within 12 month of delivery.1 Edmondson et al2 estimate the prevalence of paternal PPD to be 8% between birth and 3 months, 26% from 3 to 6 months, and 9% from 6 to 12 months.
Risk factors
Risk factors for paternal PPD have not been studied extensively. Some studies have shown that immaturity, lack of social support, first or unplanned pregnancies, marital relationship problems, and unemployment were the most common risk factors for depression in men postnatally.3 A history of depression and other psychiatric disorders also increases risk.4 Psychosocial factors, such as quality of the spousal relationship, parenting distress, and perceived parenting efficacy, contribute to paternal depression.
Similarly, depressed postpartum fathers experience higher levels of parenting distress and a lower sense of parenting efficacy.5 Interestingly, negative life events were associated with increased risk for depression in mothers, but not fathers.3
Clinical presentation
Paternal PPD symptoms appear within 12 months after the birth of the child and last for at least 2 weeks. Signs and symptoms of depression in men might not resemble those seen in postpartum women. Men tend to show aggression, increased or easy irritability, and agitation, and might not seek help for emotional issues as readily as women do. Typical symptoms of depression often are present, such as sleep disturbance or changes in sleep patterns, difficulty concentrating, memory problems, and feelings of worthlessness, hopelessness, inadequacy, and excess guilt with suicidal ideation.6
Making the diagnosis
Maternal PPD commonly is evaluated using the Edinburgh Postnatal Depression Scale- Partner (EDPS-P) or Postpartum Depression Screening Scale. However, studies are lacking to determine which diagnostic modality is most accurate for diagnosing paternal PPD.
A paternal PPD screening tool could include the EDPS-P administered to mothers. Edmonson et al2 determined an EDPS-P score of >10 was the optimal cut-off point for screening for paternal depression, with a sensitivity of 89.5% and a specificity of 78.2%, compared with a structured clinical interview. Fisher et al4 determined that the EDPS-P report was a reliable method for detecting paternal PPD compared with validated depression scales completed by fathers. Madsen et al5 determined the Gotland Male Depression Scale, which detects typical male depressive symptoms, also was effective in recognizing paternal PPD at 6 weeks postpartum.7
Treatment of paternal PPD
Specific treatment for paternal PPD has not been studied extensively. Psychotherapy targeted at interpersonal family relationships and parenting is indicated for mild depression, whereas a combination of psychotherapy and pharmacotherapy is recommended for moderate or severe depression.
Depending on specific patient factors, pharmacotherapy options include selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors, tricyclic antidepressants, and atypical antipsychotics.8 SSRIs often are used because of their efficacy and relative lack of serious side effects, as demonstrated in numerous trials.2 Recovery is more likely with combination therapy than monotherapy.9 Fathers with psychosis or suicidal ideation should be referred for inpatient treatment.
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
Only recently has paternal postpartum depression (PPD) received much attention. Research has shown that maternal PPD is associated with negative outcomes in the child’s cognitive development and social and marital problems for the parents. Likewise, depressed fathers are less likely to play outside with their child and more likely to put the child to bed awake.1
Recent studies reported that 10.4% of men experienced depression within 12 month of delivery.1 Edmondson et al2 estimate the prevalence of paternal PPD to be 8% between birth and 3 months, 26% from 3 to 6 months, and 9% from 6 to 12 months.
Risk factors
Risk factors for paternal PPD have not been studied extensively. Some studies have shown that immaturity, lack of social support, first or unplanned pregnancies, marital relationship problems, and unemployment were the most common risk factors for depression in men postnatally.3 A history of depression and other psychiatric disorders also increases risk.4 Psychosocial factors, such as quality of the spousal relationship, parenting distress, and perceived parenting efficacy, contribute to paternal depression.
Similarly, depressed postpartum fathers experience higher levels of parenting distress and a lower sense of parenting efficacy.5 Interestingly, negative life events were associated with increased risk for depression in mothers, but not fathers.3
Clinical presentation
Paternal PPD symptoms appear within 12 months after the birth of the child and last for at least 2 weeks. Signs and symptoms of depression in men might not resemble those seen in postpartum women. Men tend to show aggression, increased or easy irritability, and agitation, and might not seek help for emotional issues as readily as women do. Typical symptoms of depression often are present, such as sleep disturbance or changes in sleep patterns, difficulty concentrating, memory problems, and feelings of worthlessness, hopelessness, inadequacy, and excess guilt with suicidal ideation.6
Making the diagnosis
Maternal PPD commonly is evaluated using the Edinburgh Postnatal Depression Scale- Partner (EDPS-P) or Postpartum Depression Screening Scale. However, studies are lacking to determine which diagnostic modality is most accurate for diagnosing paternal PPD.
A paternal PPD screening tool could include the EDPS-P administered to mothers. Edmonson et al2 determined an EDPS-P score of >10 was the optimal cut-off point for screening for paternal depression, with a sensitivity of 89.5% and a specificity of 78.2%, compared with a structured clinical interview. Fisher et al4 determined that the EDPS-P report was a reliable method for detecting paternal PPD compared with validated depression scales completed by fathers. Madsen et al5 determined the Gotland Male Depression Scale, which detects typical male depressive symptoms, also was effective in recognizing paternal PPD at 6 weeks postpartum.7
Treatment of paternal PPD
Specific treatment for paternal PPD has not been studied extensively. Psychotherapy targeted at interpersonal family relationships and parenting is indicated for mild depression, whereas a combination of psychotherapy and pharmacotherapy is recommended for moderate or severe depression.
Depending on specific patient factors, pharmacotherapy options include selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors, tricyclic antidepressants, and atypical antipsychotics.8 SSRIs often are used because of their efficacy and relative lack of serious side effects, as demonstrated in numerous trials.2 Recovery is more likely with combination therapy than monotherapy.9 Fathers with psychosis or suicidal ideation should be referred for inpatient treatment.
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Paulson JF, Dauber S, Leiferman JA. Individual and combined effects of postpartum depression in mothers and fathers on parenting behavior. Pediatrics. 2006;118(2):659-668.
2. Edmondson OJ, Psychogiou L, Vlachos H, et al. Depression in fathers in the postnatal period: assessment of the Edinburgh Postnatal Depression Scale as a screening measure. J Affect Disord. 2010;125(1-3):365-368.
3. Schumacher M, Zubaran C, White G. Bringing birth-related paternal depression to the fore. Women Birth. 2008;21(2):65-70.
4. Fisher SD, Kopelman R, O’Hara MW. Partner report of paternal depression using the Edinburgh Postnatal Depression Scale-Partner. Arch Womens Ment Health. 2012;15(4):283-288.
5. Madsen SA, Juhl T. Paternal depression in the postnatal period assessed with traditional and male depression scales. Journal of Men’s Health and Gender. 2007;4(1):26-31.
6. Escribà-Agüir V, Artazcoz L. Gender differences in postpartum depression: a longitudinal cohort study. J Epidemiol Community Health. 2011;65(4):320-326.
7. Cuijpers P, van Straten A, Warmerdam L, et al. Psychotherapy versus the combination of psychotherapy and pharmacotherapy in the treatment of depression: a meta-analysis. Depress Anxiety. 2009;26(3):279-288.
8. Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 12 new-generation anti-depressants: a multiple-treatments meta-analysis. Lancet. 2009;373(9665):746-758.
9. Demontigny F, Girard ME, Lacharité C, et al. Psychosocial factors associated with paternal postnatal depression. J Affect Disord. 2013;15(150):44-49.
1. Paulson JF, Dauber S, Leiferman JA. Individual and combined effects of postpartum depression in mothers and fathers on parenting behavior. Pediatrics. 2006;118(2):659-668.
2. Edmondson OJ, Psychogiou L, Vlachos H, et al. Depression in fathers in the postnatal period: assessment of the Edinburgh Postnatal Depression Scale as a screening measure. J Affect Disord. 2010;125(1-3):365-368.
3. Schumacher M, Zubaran C, White G. Bringing birth-related paternal depression to the fore. Women Birth. 2008;21(2):65-70.
4. Fisher SD, Kopelman R, O’Hara MW. Partner report of paternal depression using the Edinburgh Postnatal Depression Scale-Partner. Arch Womens Ment Health. 2012;15(4):283-288.
5. Madsen SA, Juhl T. Paternal depression in the postnatal period assessed with traditional and male depression scales. Journal of Men’s Health and Gender. 2007;4(1):26-31.
6. Escribà-Agüir V, Artazcoz L. Gender differences in postpartum depression: a longitudinal cohort study. J Epidemiol Community Health. 2011;65(4):320-326.
7. Cuijpers P, van Straten A, Warmerdam L, et al. Psychotherapy versus the combination of psychotherapy and pharmacotherapy in the treatment of depression: a meta-analysis. Depress Anxiety. 2009;26(3):279-288.
8. Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 12 new-generation anti-depressants: a multiple-treatments meta-analysis. Lancet. 2009;373(9665):746-758.
9. Demontigny F, Girard ME, Lacharité C, et al. Psychosocial factors associated with paternal postnatal depression. J Affect Disord. 2013;15(150):44-49.
Some essentials to consider when opening a private psychiatric practice
Ah! the dream of opening private practice! Whether you’re a resident making less than minimum wage or a clinic employee seeing ever more patients, the allure is powerful. But, just because you’re whip-smart in matters of the mind, doesn’t mean you know how to run a business. To prevent your dream from succumbing to the siren’s allure, you’ll need to create a blueprint that gets you moving today, as well as prepare systems that will endure over the years.
Establish a business model
Before signing a lease or scheduling patients, think through these fundamental questions, not just from a clinical perspective but a business one as well:
• What kind of care would you like to provide? If you want to practice psychotherapy and medication, you’ll have fewer time slots to have to fill, but it may be more challenging to find patients who want and can afford psychotherapy from you as well.
• Where do you want to practice? Time spent commuting rarely produces income, so how close do you want your office to be to where you live? Being able to walk to work is wonderful, but is where you live the best location for your patients?
For example, downtown areas in big cities are good for providing a critical mass of patients, especially if you only want to manage patients’ medications. But if you want to see children and families, you should consider a location that is friendlier for them—usually more residential areas. Having a coffee shop nearby for waiting parents doesn’t hurt. If you work in a rural area, how easily can patients get to your office?
• Which hours do you want to work? Many patients will want to see you at “prime time”—before or after their work day or during the weekend. This might, not coincidentally, be when you don’t want to work. Consider whether there is room for compromise: Can you work 1 or 2 early or late days? Can you do 1 weekend day once in a while? If you want to see children, can you regularly be available after school?
• Will you accept insurance? Pros: The insurance companies will do the marketing for you; your practice will fill quickly; their checks don’t bounce; and, 98% of the time, the claims and payment process works just fine.
Cons: You will make less money per patient, in return for the higher volume of patients that are sent your way; the insurance companies won’t want to pay you more than they pay non-psychiatrists for psychotherapy; and the small amount of time that there are administrative problems can consume a disproportionate share of your sanity.
Run the numbers carefully
Next, think about the financial aspect. How much do you need to make, after you’ve paid business expenses and taxes, to be content? You might be tempted to work as many hours as possible, thinking that every hour off is an hour that you could have billed. Shifting your viewpoint from “hours lost” to “hours free” is a necessary approach to reduce burnout.
Once you have figured out your financial goal, do the math: multiply hours/ week × hourly rate × how many weeks/ year you’ll work to determine your annual income. Play around with the numbers to test your priorities, such as optimizing daily hours vs vacation time vs charging more or less.
Build your brand
This is your professional identity—the picture of your practice that your colleagues and future patients will see and that will start to get those hours filled. How will you convey your strengths and personality? The answer: Get out of the office.
• Take clinicians who will refer patients to you out to lunch (and pick up the tab).
• Give free talks to psychotherapists or primary care providers. Grand rounds, group practice meetings, or local clinical associations are potential venues. Give the organizer a menu of topic options that connect your clinical interests and theirs, and then create a dynamic presentation based on their feedback. Tip: Do not PowerPoint them to tears.
• Start blogging. If you enjoy writing, use a blog to showcase your talent and expertise. It is free advertising and makes you seem like a trusted authority. However, don’t start a blog unless you can commit to posting regularly.
Proceed thoughtfully; seek advice
As you think through the matrix of issues presented above, each set of answers may lead to a deeper set of questions. Consultation with a colleague or mentor can save you valuable time. Although you don’t have to have all the answers before you open your practice, spending time thinking through these and other issues beforehand will optimize the chance that your dream becomes a reality.
Disclosure
Dr. Braslow is the founder of Luminello.com.
Ah! the dream of opening private practice! Whether you’re a resident making less than minimum wage or a clinic employee seeing ever more patients, the allure is powerful. But, just because you’re whip-smart in matters of the mind, doesn’t mean you know how to run a business. To prevent your dream from succumbing to the siren’s allure, you’ll need to create a blueprint that gets you moving today, as well as prepare systems that will endure over the years.
Establish a business model
Before signing a lease or scheduling patients, think through these fundamental questions, not just from a clinical perspective but a business one as well:
• What kind of care would you like to provide? If you want to practice psychotherapy and medication, you’ll have fewer time slots to have to fill, but it may be more challenging to find patients who want and can afford psychotherapy from you as well.
• Where do you want to practice? Time spent commuting rarely produces income, so how close do you want your office to be to where you live? Being able to walk to work is wonderful, but is where you live the best location for your patients?
For example, downtown areas in big cities are good for providing a critical mass of patients, especially if you only want to manage patients’ medications. But if you want to see children and families, you should consider a location that is friendlier for them—usually more residential areas. Having a coffee shop nearby for waiting parents doesn’t hurt. If you work in a rural area, how easily can patients get to your office?
• Which hours do you want to work? Many patients will want to see you at “prime time”—before or after their work day or during the weekend. This might, not coincidentally, be when you don’t want to work. Consider whether there is room for compromise: Can you work 1 or 2 early or late days? Can you do 1 weekend day once in a while? If you want to see children, can you regularly be available after school?
• Will you accept insurance? Pros: The insurance companies will do the marketing for you; your practice will fill quickly; their checks don’t bounce; and, 98% of the time, the claims and payment process works just fine.
Cons: You will make less money per patient, in return for the higher volume of patients that are sent your way; the insurance companies won’t want to pay you more than they pay non-psychiatrists for psychotherapy; and the small amount of time that there are administrative problems can consume a disproportionate share of your sanity.
Run the numbers carefully
Next, think about the financial aspect. How much do you need to make, after you’ve paid business expenses and taxes, to be content? You might be tempted to work as many hours as possible, thinking that every hour off is an hour that you could have billed. Shifting your viewpoint from “hours lost” to “hours free” is a necessary approach to reduce burnout.
Once you have figured out your financial goal, do the math: multiply hours/ week × hourly rate × how many weeks/ year you’ll work to determine your annual income. Play around with the numbers to test your priorities, such as optimizing daily hours vs vacation time vs charging more or less.
Build your brand
This is your professional identity—the picture of your practice that your colleagues and future patients will see and that will start to get those hours filled. How will you convey your strengths and personality? The answer: Get out of the office.
• Take clinicians who will refer patients to you out to lunch (and pick up the tab).
• Give free talks to psychotherapists or primary care providers. Grand rounds, group practice meetings, or local clinical associations are potential venues. Give the organizer a menu of topic options that connect your clinical interests and theirs, and then create a dynamic presentation based on their feedback. Tip: Do not PowerPoint them to tears.
• Start blogging. If you enjoy writing, use a blog to showcase your talent and expertise. It is free advertising and makes you seem like a trusted authority. However, don’t start a blog unless you can commit to posting regularly.
Proceed thoughtfully; seek advice
As you think through the matrix of issues presented above, each set of answers may lead to a deeper set of questions. Consultation with a colleague or mentor can save you valuable time. Although you don’t have to have all the answers before you open your practice, spending time thinking through these and other issues beforehand will optimize the chance that your dream becomes a reality.
Disclosure
Dr. Braslow is the founder of Luminello.com.
Ah! the dream of opening private practice! Whether you’re a resident making less than minimum wage or a clinic employee seeing ever more patients, the allure is powerful. But, just because you’re whip-smart in matters of the mind, doesn’t mean you know how to run a business. To prevent your dream from succumbing to the siren’s allure, you’ll need to create a blueprint that gets you moving today, as well as prepare systems that will endure over the years.
Establish a business model
Before signing a lease or scheduling patients, think through these fundamental questions, not just from a clinical perspective but a business one as well:
• What kind of care would you like to provide? If you want to practice psychotherapy and medication, you’ll have fewer time slots to have to fill, but it may be more challenging to find patients who want and can afford psychotherapy from you as well.
• Where do you want to practice? Time spent commuting rarely produces income, so how close do you want your office to be to where you live? Being able to walk to work is wonderful, but is where you live the best location for your patients?
For example, downtown areas in big cities are good for providing a critical mass of patients, especially if you only want to manage patients’ medications. But if you want to see children and families, you should consider a location that is friendlier for them—usually more residential areas. Having a coffee shop nearby for waiting parents doesn’t hurt. If you work in a rural area, how easily can patients get to your office?
• Which hours do you want to work? Many patients will want to see you at “prime time”—before or after their work day or during the weekend. This might, not coincidentally, be when you don’t want to work. Consider whether there is room for compromise: Can you work 1 or 2 early or late days? Can you do 1 weekend day once in a while? If you want to see children, can you regularly be available after school?
• Will you accept insurance? Pros: The insurance companies will do the marketing for you; your practice will fill quickly; their checks don’t bounce; and, 98% of the time, the claims and payment process works just fine.
Cons: You will make less money per patient, in return for the higher volume of patients that are sent your way; the insurance companies won’t want to pay you more than they pay non-psychiatrists for psychotherapy; and the small amount of time that there are administrative problems can consume a disproportionate share of your sanity.
Run the numbers carefully
Next, think about the financial aspect. How much do you need to make, after you’ve paid business expenses and taxes, to be content? You might be tempted to work as many hours as possible, thinking that every hour off is an hour that you could have billed. Shifting your viewpoint from “hours lost” to “hours free” is a necessary approach to reduce burnout.
Once you have figured out your financial goal, do the math: multiply hours/ week × hourly rate × how many weeks/ year you’ll work to determine your annual income. Play around with the numbers to test your priorities, such as optimizing daily hours vs vacation time vs charging more or less.
Build your brand
This is your professional identity—the picture of your practice that your colleagues and future patients will see and that will start to get those hours filled. How will you convey your strengths and personality? The answer: Get out of the office.
• Take clinicians who will refer patients to you out to lunch (and pick up the tab).
• Give free talks to psychotherapists or primary care providers. Grand rounds, group practice meetings, or local clinical associations are potential venues. Give the organizer a menu of topic options that connect your clinical interests and theirs, and then create a dynamic presentation based on their feedback. Tip: Do not PowerPoint them to tears.
• Start blogging. If you enjoy writing, use a blog to showcase your talent and expertise. It is free advertising and makes you seem like a trusted authority. However, don’t start a blog unless you can commit to posting regularly.
Proceed thoughtfully; seek advice
As you think through the matrix of issues presented above, each set of answers may lead to a deeper set of questions. Consultation with a colleague or mentor can save you valuable time. Although you don’t have to have all the answers before you open your practice, spending time thinking through these and other issues beforehand will optimize the chance that your dream becomes a reality.
Disclosure
Dr. Braslow is the founder of Luminello.com.
Sober today, but lethargic and confused
CASE Confused and weak
Mr. W, age 26, is brought to the emergency department (ED) by his parents for intermittent confusion, weakness, and increasing lethargy over the past 4 days. He is jaundiced with mild abdominal pain, nausea, and vomiting.
Mr. W has a history of alcohol use disorder, drinking as much as 1 L of vodka a day. Six months ago, he was hospitalized for alcoholic hepatitis and severe hyponatremia.
In the ED, Mr. W is awake, alert, and oriented to person, place, and time. Vital signs are: pulse 89 beats per minute; blood pressure, 117/50 mm Hg; respirations, 15 breaths per minute; and temperature, 98.5ºF. Physical examination is notable for scleral icterus, jaundice, tender hepatomegaly, and asterixis.
Mr. W is not taking any medications. He reports that his most recent drink was the day before; however, his current alcohol intake is unknown.
Laboratory tests reveal altered hepatic function, including elevated aspartate aminotransferase (251 U/L), alanine aminotransferase (56 U/L), alkaline phosphatase (179 U/L), total bilirubin (15.4 mg/dL), and ammonia (143 U/L), impaired coagulation (international normalized ratio 2.39), and decreased albumin (2.7 g/dL). Other metabolic disturbances include: sodium, 104 mEq/L; chloride, <60 mEq/L; potassium, 2.2 mEq/L; and CO2, 44.5 mEq/L.
What is your differential diagnosis for Mr. W’s altered mental status?
a) hepatic encephalopathy
b) Wernicke’s encephalopathy
c) hyponatremia
d) drug intoxication
e) head trauma
The authors’ observations
Hyponatremia is defined as a serum sodium concentration <136 mEq/L. Mr. W is considered to have severe hyponatremia because his serum sodium concentration is <125 mEq/L. Although commonly caused by an inability to suppress antidiuretic hormone, hyponatremia has several possible causes (Figure 1).1 Symptoms are nonspecific and are more visible when there is a large or rapid decrease in the serum sodium concentration. Most patients with a serum sodium concentration >125 mEq/L are asymptomatic. Mr. W, who had a serum sodium of 104 mEq/L, presented with several symptoms, including confusion, lethargy, nausea, vomiting, and weakness. Headache, muscle spasms, depressed reflexes, restlessness, and disorientation also might be observed.2
Because of Mr. W’s impaired hepatic function, elevated ammonia, and asterixis, hepatic encephalopathy could be contributing to his altered mental status. Suspect Wernicke’s encephalopathy in a patient with neurologic symptoms and a history of chronic alcohol abuse. In its classic form, Wernicke’s encephalopathy has acute onset, characterized by the triad of ataxia, global confusion, and ocular abnormalities. However, this triad is not consistently or frequently encountered.3
Which tests would you order next?
a) blood ethanol level
b) urine drug screen
c) serum osmolality
d) CT of the head
EVALUATION Sober, yet sick
To rule out intoxication as the cause of Mr. W’s altered mental status, blood ethanol level and urine drug screens are obtained and found to be negative. CT of the head is negative for acute intracranial pathology.
Mr. W is admitted to the medical intensive care unit (MICU) for severe hyponatremia and altered mental status. Serum osmolality is 220 mOsm/kg (normal range 281 to 304 mOsm/kg). To further classify his hypotonic hyponatremia, volume status is assessed, and Mr. W is determined to be euvolemic. Thyroid-stimulating hormone and cortisol are within normal limits, eliminating hypothyroidism and adrenal insufficiency as causes of his euvolemic hypotonic hyponatremia. Mr. W is treated for hyponatremia likely secondary to syndrome of inappropriate antidiuretic hormone (SIADH). SIADH is a diagnosis of exclusion that first requires ruling out hypothyroidism and glucocorticoid insufficiency (Figure 1).1
The authors’ observations
Because hypokalemia is an independent predictive factor for development of hyponatremia, it is necessary to evaluate the potassium level in all hyponatremic patients. Mr. W’s potassium level was 2.2 mEq/L on admission. Serum sodium concentration is related to total exchangeable sodium, total body water, and total exchangeable potassium. Potassium depletion causes a shift of sodium into cells with a comparable exit of potassium from cells into extracellular fluid. The reverse process occurs during potassium repletion, leading to an increase in serum sodium concentration and making hypokalemia a risk factor for developing osmotic demyelination syndrome (ODS).4
Treating hyponatremia
Hyponatremia treatment depends on its severity, presence or absence of symptoms, and whether the hyponatremia is acute (<24 hours) or chronic (>48 hours).5
Because of Mr. W’s extremely low serum sodium concentration, predisposition to hyponatremia secondary to alcoholism, and history of severe hyponatremia, it is likely he is chronically hyponatremic.
In patients with chronic hyponatremia, neurological sequelae are associated with the need for a more rapid rate of correction of serum sodium. For most patients with chronic hyponatremia, a correction rate of ≤10 to 12 mEq/L during the first 24 hours and <18 mEq/L over 48 hours is recommended to avoid ODS.6
Evidence suggests, however, that this 1-day limit might be too high for some patients. Alcoholism, hypokalemia, malnutrition, and liver disease are present in a high percentage of patients who develop
ODS after correcting hyponatremia (Table 1).6 Therefore, for patients such as Mr. W who are at high risk of ODS, experts recommend a goal of 4 to 6 mEq/L/d with a correction rate of ≤8 mEq/L in any 24-hour period (Table 2).6
TREATMENT Sodium normalizes
Mr. W receives 1 L of normal saline in the ED before admission to the MICU. Once in the MICU, despite likely chronic hyponatremia, he receives hypertonic (3%) saline, followed by normal saline. Initially, he responds when the serum sodium concentration improves. Because of his likely SIADH, Mr. W is fluid-restricted for 4 days. Serum sodium returns to normal over 7 hospital days (Figure 2). To address the profound hypokalemia, Mr. S receives 30 mEq of potassium chloride in the ED, and potassium is repeated daily throughout his stay in the MICU.
Mr. W remains lethargic, with intermittent periods of confusion throughout the hospital stay. His altered mental status is attributed to hepatic encephalopathy secondary to alcoholic hepatitis. The Maddrey discriminant function is a calculation that stratifies patients with alcoholic hepatitis for risk of mortality and the use of steroids. Because Mr. W shows a Maddrey discriminant function ≥32, he receives methylprednisolone, followed by pentoxifylline, and liver function tests trend down. He also receives lactulose throughout hospitalization.
By discharge on hospital day 9, Mr. W’s serum sodium is 138 mEq/L; serum potassium, 4.1 mEq/L. Total bilirubin and prothrombin remain elevated. Mr. W is discharged on lactulose, thiamine, folic acid, and a 1-month course of pentoxifylline, 400 mg, 3 times a day.
READMISSION Unsteady gait, nausea
Three days after discharge, Mr. W returns to the ED after experiencing a 20-second episode of total body rigidity. He has an unsteady gait and worsening nausea and vomiting.
When Mr. W arrives in the ED, he confirms he is taking his discharge medications as prescribed. His parents report that he has consumed alcohol and Cannabis since discharge and has been taking his sibling’s prescription medications, including quetiapine.
In the ED, Mr. W is awake, alert, and oriented to person, place, and time. Vital signs are: pulse, 118 beats per minute; blood pressure, 128/73 mm Hg; respirations, 16 breaths per minute; and temperature, 98.5ºF. Physical examination, again, is notable for scleral icterus, jaundice, and asterixis. No focal neurologic deficits are noted.
Consistent with Mr. W’s previous admission, laboratory values reveal altered hepatic function and impaired coagulation. The serum sodium level remains within normal limits at 136 mEq/L. However, again, metabolic disturbances include decreased chloride (97 mEq/L), potassium (2.9 mEq/L), and CO2 (18.2 mEq/L). CT on readmission is unchanged from the earlier hospitalization.
What is your differential diagnosis for Mr. W’s total body rigidity?
a) seizure
b) ODS
c) drug intoxication
d) neuroleptic malignant syndrome
EVALUATION Shaking and weakness
Once admitted to the hospital, Mr. W reports an episode of right upper-extremity “shaking,” followed by weakness. He remembers the entire event and denies tongue biting or incontinence. He is evaluated for possible seizure, given his multiple risk factors, including drug and alcohol use, ingestion of quetiapine, and history of hyponatremia. Routine EEG is negative but prolactin level is elevated.
Mr. W’s mental status continues to wax and wane, prompting a neurology consult and MRI for further evaluation. MRI of the brain without contrast reveals restricted diffusion in the pons centrally, with extension bilaterally to the midbrain and thalami—findings consistent with central pontine myelinolysis. A neurology consultation reveals quadriparesis, paraparesis, dysarthria, and diplopia on examination, all symptoms associated with central pontine myelinolysis.
The authors’ observations
ODS, including central and extrapontine myelinolysis, is a demyelinating condition that occurs because of severe osmotic stress, most commonly secondary to the overly rapid correction of hyponatremia in patients with conditions leading to nutritional or electrolyte stress.7 Mr. W is considered at high risk of developing ODS because he fulfills the 5 criteria listed in Table 1.
Several psychiatric illnesses and neuropsychiatric medications could lead to hyponatremia. Many studies8-10 have documented hyponatremia and resulting ODS in patients with alcoholism, schizophrenia, anorexia, primary psychogenic polydipsia, and MDMA (3,4-methylenedioxymethamphetamine) abuse. Hyponatremia is a side effect of several neuropsychiatric medications, including serotonin reuptake inhibitors, lithium, tricyclic antidepressants, opioids, carbamazepine, oxcarbazepine, and antipsychotic polypharmacy. Other commonly used medications associated with hyponatremia include salt-losing diuretics, nonsteroidal anti-inflammatory drugs, and acetaminophen.7
Disease severity varies from asymptomatic to coma or death. Symptoms, although some could reverse completely, typically are a combination of neuropsychiatric (ie, emotional lability, disinhibition, and other bizarre behaviors) and neurologic. Neurologic symptoms include confusion, impaired cognition, dysarthria, dysphagia, gait instability, weakness or paralysis, and generalized seizures. Severely affected patients could experience “locked-in syndrome,” in which they are awake but unable to move or communicate. Also consistent with Mr. W’s case, ODS often presents initially with delirium, seizures, or encephalopathy, followed by a lucid interval before symptoms develop.7
Diagnosis is based on the appearance of demyelinating brain lesions on CT or MRI. MRI is more sensitive than CT; however, even an MRI scan can appear normal for as long as 4 weeks after symptoms appear.7 Therefore, an initial negative radiologic study in a high-risk patient who develops neurologic symptoms does not exclude ODS. Earlier detection is possible with diffusion-weighted MRI, which is most sensitive and can detect lesions within 24 hours of developing symptoms.11 The severity of the lesion does not correlate with severity of symptoms.
Studies reveal a considerable range in prognosis of patients with clinically symptomatic ODS. A study of 44 patients with central pontine myelinolysis, of which 42 had chronic alcoholism, reported that 34% had no significant functional deficits at follow-up, 34% had minor neurologic deficits, and 31% became dependent on personal help. Outcome did not depend on the extent or severity of neurologic symptoms or the severity of concomitant systemic complications.12
Because of its poor prognosis, prevention of ODS is important. Because ODS commonly is caused by overly rapid correction of hyponatremia, it is necessary to adhere to guidelines for treating chronic hyponatremia (Table 2). If overcorrection occurs, therapeutic re-lowering of serum sodium can be considered, but has not been validated in controlled trials. Based mainly on case reports that suggest benefit from early re-lowering serum sodium in patients with ODS symptoms, experts recommend the following:
• administer desmopressin, 2 to 4 μg, every 8 hours parenterally
• replace water orally or as 5% dextrose in water intravenously (3 mL/kg/hr)
• check serum sodium hourly until serum is reduced to goal.6
Bottom Line
Hyponatremia is the most common electrolyte disorder encountered in practice. Osmotic demyelination syndrome often is preventable, with considerable morbidity and mortality. Psychiatrists should be aware of this condition because it could be an adverse effect of many psychiatric medications and there are some psychiatric illnesses in which hyponatremia is a potential risk. In hyponatremic patients with persistent nonspecific neurologic or neuropsychiatric symptoms and negative CT imaging, additional imaging, such as MRI, is warranted.
Related Resources
- Braun MM, Barstow CH, Pyzocha NJ. Diagnosis and management of sodium disorders: hyponatremia and hypernatremia. Am Fam Physician. 2015;91(5):299-307.
- Vaidya C, Ho W, Freda BJ. Management of hyponatremia: providing treatment and avoiding harm. Cleve Clin J Med. 2010;77(10):715-726.
Drug Brand Names
Carbamazepine • Tegretol
Oxcarbazepine • Trileptal
Desmopressin • Stimate, DDAVP
Lithium • Eskalith, Lithobid
Pentoxifylline • Trental, Pentoxil
Methylprednisolone • Medrol
Quetiapine • Seroquel
Disclosure
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Elhassen EA, Schrier RW. Disorders of sodium and water balance. In: McKean SC, Ross JJ, Dressler DD, et al, eds. Principles and practice of hospital medicine. New York, NY: McGraw-Hill; 2012:2084-2093.
2. Adrogué HJ, Madias NE. Hyponatremia. N Engl J Med. 2000;342(21):1581-1589.
3. Reuler JB, Girard DE, Cooney TG. Current concepts. Wernicke’s encephalopathy. N Engl J Med. 1985;312(16):1035-1039.
4. Edelman IS, Leibman J, O’Meara MP, et al. Interrelations between serum sodium concentration, serum osmolarity and total exchangeable sodium, total exchangeable potassium and total body water. J Clin Invest. 1958;37(9):1236-1256.
5. Reynolds RM, Seckl JR. Hyponatraemia for the clinical endocrinologist. Clin Endocrinol (Oxf). 2005;63(4):366-374.
6. Verbalis JG, Goldsmith SR, Greenberg A, et al. Diagnosis, evaluation, and treatment of hyponatremia: expert panel recommendations. Am J Med. 2013;126(10 suppl 1):S1-S42.
7. Hurley RA, Filley CM, Taber KH. Central pontine myelinolysis: a metabolic disorder of myelin. J Neuropsychiatry Clin Neurosci. 2011;23(4):369-374.
8. Goldman MB. The assessment and treatment of water imbalance in patients with psychosis. Clin Schizophr Related Psychoses. 2010;4(2):115-123.
9. Patel AS, Matthews L, Bruce-Jones W. Central pontine myelinolysis as a complication of refeeding syndrome in a patient with anorexia nervosa. J Neuropsychiatry Clin Neurosci. 2008;20(3):371-373.
10. Bhuvaneswar CG, Baldessarini RJ, Harsh VL, et al. Adverse endocrine and metabolic effects of psychotropic drugs: selective clinical review. CNS Drugs. 2009;23(12):1003-1021.
11. Ruzek KA, Campeau NG, Miller GM. Early diagnosis of central pontine myelinolysis with diffusion-weighted imaging. AJNR Am J Neuroradiol. 2004;25(2):210-213.
12. Menger H, Jörg J. Outcome of central pontine and extrapontine myelinolysis (n = 44). J Neurol. 1999;246(8):700-705.
sodium concentration, Wernicke’s
encephalopathy, osmotic demyelination syndrome, electrolyte disorder
CASE Confused and weak
Mr. W, age 26, is brought to the emergency department (ED) by his parents for intermittent confusion, weakness, and increasing lethargy over the past 4 days. He is jaundiced with mild abdominal pain, nausea, and vomiting.
Mr. W has a history of alcohol use disorder, drinking as much as 1 L of vodka a day. Six months ago, he was hospitalized for alcoholic hepatitis and severe hyponatremia.
In the ED, Mr. W is awake, alert, and oriented to person, place, and time. Vital signs are: pulse 89 beats per minute; blood pressure, 117/50 mm Hg; respirations, 15 breaths per minute; and temperature, 98.5ºF. Physical examination is notable for scleral icterus, jaundice, tender hepatomegaly, and asterixis.
Mr. W is not taking any medications. He reports that his most recent drink was the day before; however, his current alcohol intake is unknown.
Laboratory tests reveal altered hepatic function, including elevated aspartate aminotransferase (251 U/L), alanine aminotransferase (56 U/L), alkaline phosphatase (179 U/L), total bilirubin (15.4 mg/dL), and ammonia (143 U/L), impaired coagulation (international normalized ratio 2.39), and decreased albumin (2.7 g/dL). Other metabolic disturbances include: sodium, 104 mEq/L; chloride, <60 mEq/L; potassium, 2.2 mEq/L; and CO2, 44.5 mEq/L.
What is your differential diagnosis for Mr. W’s altered mental status?
a) hepatic encephalopathy
b) Wernicke’s encephalopathy
c) hyponatremia
d) drug intoxication
e) head trauma
The authors’ observations
Hyponatremia is defined as a serum sodium concentration <136 mEq/L. Mr. W is considered to have severe hyponatremia because his serum sodium concentration is <125 mEq/L. Although commonly caused by an inability to suppress antidiuretic hormone, hyponatremia has several possible causes (Figure 1).1 Symptoms are nonspecific and are more visible when there is a large or rapid decrease in the serum sodium concentration. Most patients with a serum sodium concentration >125 mEq/L are asymptomatic. Mr. W, who had a serum sodium of 104 mEq/L, presented with several symptoms, including confusion, lethargy, nausea, vomiting, and weakness. Headache, muscle spasms, depressed reflexes, restlessness, and disorientation also might be observed.2
Because of Mr. W’s impaired hepatic function, elevated ammonia, and asterixis, hepatic encephalopathy could be contributing to his altered mental status. Suspect Wernicke’s encephalopathy in a patient with neurologic symptoms and a history of chronic alcohol abuse. In its classic form, Wernicke’s encephalopathy has acute onset, characterized by the triad of ataxia, global confusion, and ocular abnormalities. However, this triad is not consistently or frequently encountered.3
Which tests would you order next?
a) blood ethanol level
b) urine drug screen
c) serum osmolality
d) CT of the head
EVALUATION Sober, yet sick
To rule out intoxication as the cause of Mr. W’s altered mental status, blood ethanol level and urine drug screens are obtained and found to be negative. CT of the head is negative for acute intracranial pathology.
Mr. W is admitted to the medical intensive care unit (MICU) for severe hyponatremia and altered mental status. Serum osmolality is 220 mOsm/kg (normal range 281 to 304 mOsm/kg). To further classify his hypotonic hyponatremia, volume status is assessed, and Mr. W is determined to be euvolemic. Thyroid-stimulating hormone and cortisol are within normal limits, eliminating hypothyroidism and adrenal insufficiency as causes of his euvolemic hypotonic hyponatremia. Mr. W is treated for hyponatremia likely secondary to syndrome of inappropriate antidiuretic hormone (SIADH). SIADH is a diagnosis of exclusion that first requires ruling out hypothyroidism and glucocorticoid insufficiency (Figure 1).1
The authors’ observations
Because hypokalemia is an independent predictive factor for development of hyponatremia, it is necessary to evaluate the potassium level in all hyponatremic patients. Mr. W’s potassium level was 2.2 mEq/L on admission. Serum sodium concentration is related to total exchangeable sodium, total body water, and total exchangeable potassium. Potassium depletion causes a shift of sodium into cells with a comparable exit of potassium from cells into extracellular fluid. The reverse process occurs during potassium repletion, leading to an increase in serum sodium concentration and making hypokalemia a risk factor for developing osmotic demyelination syndrome (ODS).4
Treating hyponatremia
Hyponatremia treatment depends on its severity, presence or absence of symptoms, and whether the hyponatremia is acute (<24 hours) or chronic (>48 hours).5
Because of Mr. W’s extremely low serum sodium concentration, predisposition to hyponatremia secondary to alcoholism, and history of severe hyponatremia, it is likely he is chronically hyponatremic.
In patients with chronic hyponatremia, neurological sequelae are associated with the need for a more rapid rate of correction of serum sodium. For most patients with chronic hyponatremia, a correction rate of ≤10 to 12 mEq/L during the first 24 hours and <18 mEq/L over 48 hours is recommended to avoid ODS.6
Evidence suggests, however, that this 1-day limit might be too high for some patients. Alcoholism, hypokalemia, malnutrition, and liver disease are present in a high percentage of patients who develop
ODS after correcting hyponatremia (Table 1).6 Therefore, for patients such as Mr. W who are at high risk of ODS, experts recommend a goal of 4 to 6 mEq/L/d with a correction rate of ≤8 mEq/L in any 24-hour period (Table 2).6
TREATMENT Sodium normalizes
Mr. W receives 1 L of normal saline in the ED before admission to the MICU. Once in the MICU, despite likely chronic hyponatremia, he receives hypertonic (3%) saline, followed by normal saline. Initially, he responds when the serum sodium concentration improves. Because of his likely SIADH, Mr. W is fluid-restricted for 4 days. Serum sodium returns to normal over 7 hospital days (Figure 2). To address the profound hypokalemia, Mr. S receives 30 mEq of potassium chloride in the ED, and potassium is repeated daily throughout his stay in the MICU.
Mr. W remains lethargic, with intermittent periods of confusion throughout the hospital stay. His altered mental status is attributed to hepatic encephalopathy secondary to alcoholic hepatitis. The Maddrey discriminant function is a calculation that stratifies patients with alcoholic hepatitis for risk of mortality and the use of steroids. Because Mr. W shows a Maddrey discriminant function ≥32, he receives methylprednisolone, followed by pentoxifylline, and liver function tests trend down. He also receives lactulose throughout hospitalization.
By discharge on hospital day 9, Mr. W’s serum sodium is 138 mEq/L; serum potassium, 4.1 mEq/L. Total bilirubin and prothrombin remain elevated. Mr. W is discharged on lactulose, thiamine, folic acid, and a 1-month course of pentoxifylline, 400 mg, 3 times a day.
READMISSION Unsteady gait, nausea
Three days after discharge, Mr. W returns to the ED after experiencing a 20-second episode of total body rigidity. He has an unsteady gait and worsening nausea and vomiting.
When Mr. W arrives in the ED, he confirms he is taking his discharge medications as prescribed. His parents report that he has consumed alcohol and Cannabis since discharge and has been taking his sibling’s prescription medications, including quetiapine.
In the ED, Mr. W is awake, alert, and oriented to person, place, and time. Vital signs are: pulse, 118 beats per minute; blood pressure, 128/73 mm Hg; respirations, 16 breaths per minute; and temperature, 98.5ºF. Physical examination, again, is notable for scleral icterus, jaundice, and asterixis. No focal neurologic deficits are noted.
Consistent with Mr. W’s previous admission, laboratory values reveal altered hepatic function and impaired coagulation. The serum sodium level remains within normal limits at 136 mEq/L. However, again, metabolic disturbances include decreased chloride (97 mEq/L), potassium (2.9 mEq/L), and CO2 (18.2 mEq/L). CT on readmission is unchanged from the earlier hospitalization.
What is your differential diagnosis for Mr. W’s total body rigidity?
a) seizure
b) ODS
c) drug intoxication
d) neuroleptic malignant syndrome
EVALUATION Shaking and weakness
Once admitted to the hospital, Mr. W reports an episode of right upper-extremity “shaking,” followed by weakness. He remembers the entire event and denies tongue biting or incontinence. He is evaluated for possible seizure, given his multiple risk factors, including drug and alcohol use, ingestion of quetiapine, and history of hyponatremia. Routine EEG is negative but prolactin level is elevated.
Mr. W’s mental status continues to wax and wane, prompting a neurology consult and MRI for further evaluation. MRI of the brain without contrast reveals restricted diffusion in the pons centrally, with extension bilaterally to the midbrain and thalami—findings consistent with central pontine myelinolysis. A neurology consultation reveals quadriparesis, paraparesis, dysarthria, and diplopia on examination, all symptoms associated with central pontine myelinolysis.
The authors’ observations
ODS, including central and extrapontine myelinolysis, is a demyelinating condition that occurs because of severe osmotic stress, most commonly secondary to the overly rapid correction of hyponatremia in patients with conditions leading to nutritional or electrolyte stress.7 Mr. W is considered at high risk of developing ODS because he fulfills the 5 criteria listed in Table 1.
Several psychiatric illnesses and neuropsychiatric medications could lead to hyponatremia. Many studies8-10 have documented hyponatremia and resulting ODS in patients with alcoholism, schizophrenia, anorexia, primary psychogenic polydipsia, and MDMA (3,4-methylenedioxymethamphetamine) abuse. Hyponatremia is a side effect of several neuropsychiatric medications, including serotonin reuptake inhibitors, lithium, tricyclic antidepressants, opioids, carbamazepine, oxcarbazepine, and antipsychotic polypharmacy. Other commonly used medications associated with hyponatremia include salt-losing diuretics, nonsteroidal anti-inflammatory drugs, and acetaminophen.7
Disease severity varies from asymptomatic to coma or death. Symptoms, although some could reverse completely, typically are a combination of neuropsychiatric (ie, emotional lability, disinhibition, and other bizarre behaviors) and neurologic. Neurologic symptoms include confusion, impaired cognition, dysarthria, dysphagia, gait instability, weakness or paralysis, and generalized seizures. Severely affected patients could experience “locked-in syndrome,” in which they are awake but unable to move or communicate. Also consistent with Mr. W’s case, ODS often presents initially with delirium, seizures, or encephalopathy, followed by a lucid interval before symptoms develop.7
Diagnosis is based on the appearance of demyelinating brain lesions on CT or MRI. MRI is more sensitive than CT; however, even an MRI scan can appear normal for as long as 4 weeks after symptoms appear.7 Therefore, an initial negative radiologic study in a high-risk patient who develops neurologic symptoms does not exclude ODS. Earlier detection is possible with diffusion-weighted MRI, which is most sensitive and can detect lesions within 24 hours of developing symptoms.11 The severity of the lesion does not correlate with severity of symptoms.
Studies reveal a considerable range in prognosis of patients with clinically symptomatic ODS. A study of 44 patients with central pontine myelinolysis, of which 42 had chronic alcoholism, reported that 34% had no significant functional deficits at follow-up, 34% had minor neurologic deficits, and 31% became dependent on personal help. Outcome did not depend on the extent or severity of neurologic symptoms or the severity of concomitant systemic complications.12
Because of its poor prognosis, prevention of ODS is important. Because ODS commonly is caused by overly rapid correction of hyponatremia, it is necessary to adhere to guidelines for treating chronic hyponatremia (Table 2). If overcorrection occurs, therapeutic re-lowering of serum sodium can be considered, but has not been validated in controlled trials. Based mainly on case reports that suggest benefit from early re-lowering serum sodium in patients with ODS symptoms, experts recommend the following:
• administer desmopressin, 2 to 4 μg, every 8 hours parenterally
• replace water orally or as 5% dextrose in water intravenously (3 mL/kg/hr)
• check serum sodium hourly until serum is reduced to goal.6
Bottom Line
Hyponatremia is the most common electrolyte disorder encountered in practice. Osmotic demyelination syndrome often is preventable, with considerable morbidity and mortality. Psychiatrists should be aware of this condition because it could be an adverse effect of many psychiatric medications and there are some psychiatric illnesses in which hyponatremia is a potential risk. In hyponatremic patients with persistent nonspecific neurologic or neuropsychiatric symptoms and negative CT imaging, additional imaging, such as MRI, is warranted.
Related Resources
- Braun MM, Barstow CH, Pyzocha NJ. Diagnosis and management of sodium disorders: hyponatremia and hypernatremia. Am Fam Physician. 2015;91(5):299-307.
- Vaidya C, Ho W, Freda BJ. Management of hyponatremia: providing treatment and avoiding harm. Cleve Clin J Med. 2010;77(10):715-726.
Drug Brand Names
Carbamazepine • Tegretol
Oxcarbazepine • Trileptal
Desmopressin • Stimate, DDAVP
Lithium • Eskalith, Lithobid
Pentoxifylline • Trental, Pentoxil
Methylprednisolone • Medrol
Quetiapine • Seroquel
Disclosure
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
CASE Confused and weak
Mr. W, age 26, is brought to the emergency department (ED) by his parents for intermittent confusion, weakness, and increasing lethargy over the past 4 days. He is jaundiced with mild abdominal pain, nausea, and vomiting.
Mr. W has a history of alcohol use disorder, drinking as much as 1 L of vodka a day. Six months ago, he was hospitalized for alcoholic hepatitis and severe hyponatremia.
In the ED, Mr. W is awake, alert, and oriented to person, place, and time. Vital signs are: pulse 89 beats per minute; blood pressure, 117/50 mm Hg; respirations, 15 breaths per minute; and temperature, 98.5ºF. Physical examination is notable for scleral icterus, jaundice, tender hepatomegaly, and asterixis.
Mr. W is not taking any medications. He reports that his most recent drink was the day before; however, his current alcohol intake is unknown.
Laboratory tests reveal altered hepatic function, including elevated aspartate aminotransferase (251 U/L), alanine aminotransferase (56 U/L), alkaline phosphatase (179 U/L), total bilirubin (15.4 mg/dL), and ammonia (143 U/L), impaired coagulation (international normalized ratio 2.39), and decreased albumin (2.7 g/dL). Other metabolic disturbances include: sodium, 104 mEq/L; chloride, <60 mEq/L; potassium, 2.2 mEq/L; and CO2, 44.5 mEq/L.
What is your differential diagnosis for Mr. W’s altered mental status?
a) hepatic encephalopathy
b) Wernicke’s encephalopathy
c) hyponatremia
d) drug intoxication
e) head trauma
The authors’ observations
Hyponatremia is defined as a serum sodium concentration <136 mEq/L. Mr. W is considered to have severe hyponatremia because his serum sodium concentration is <125 mEq/L. Although commonly caused by an inability to suppress antidiuretic hormone, hyponatremia has several possible causes (Figure 1).1 Symptoms are nonspecific and are more visible when there is a large or rapid decrease in the serum sodium concentration. Most patients with a serum sodium concentration >125 mEq/L are asymptomatic. Mr. W, who had a serum sodium of 104 mEq/L, presented with several symptoms, including confusion, lethargy, nausea, vomiting, and weakness. Headache, muscle spasms, depressed reflexes, restlessness, and disorientation also might be observed.2
Because of Mr. W’s impaired hepatic function, elevated ammonia, and asterixis, hepatic encephalopathy could be contributing to his altered mental status. Suspect Wernicke’s encephalopathy in a patient with neurologic symptoms and a history of chronic alcohol abuse. In its classic form, Wernicke’s encephalopathy has acute onset, characterized by the triad of ataxia, global confusion, and ocular abnormalities. However, this triad is not consistently or frequently encountered.3
Which tests would you order next?
a) blood ethanol level
b) urine drug screen
c) serum osmolality
d) CT of the head
EVALUATION Sober, yet sick
To rule out intoxication as the cause of Mr. W’s altered mental status, blood ethanol level and urine drug screens are obtained and found to be negative. CT of the head is negative for acute intracranial pathology.
Mr. W is admitted to the medical intensive care unit (MICU) for severe hyponatremia and altered mental status. Serum osmolality is 220 mOsm/kg (normal range 281 to 304 mOsm/kg). To further classify his hypotonic hyponatremia, volume status is assessed, and Mr. W is determined to be euvolemic. Thyroid-stimulating hormone and cortisol are within normal limits, eliminating hypothyroidism and adrenal insufficiency as causes of his euvolemic hypotonic hyponatremia. Mr. W is treated for hyponatremia likely secondary to syndrome of inappropriate antidiuretic hormone (SIADH). SIADH is a diagnosis of exclusion that first requires ruling out hypothyroidism and glucocorticoid insufficiency (Figure 1).1
The authors’ observations
Because hypokalemia is an independent predictive factor for development of hyponatremia, it is necessary to evaluate the potassium level in all hyponatremic patients. Mr. W’s potassium level was 2.2 mEq/L on admission. Serum sodium concentration is related to total exchangeable sodium, total body water, and total exchangeable potassium. Potassium depletion causes a shift of sodium into cells with a comparable exit of potassium from cells into extracellular fluid. The reverse process occurs during potassium repletion, leading to an increase in serum sodium concentration and making hypokalemia a risk factor for developing osmotic demyelination syndrome (ODS).4
Treating hyponatremia
Hyponatremia treatment depends on its severity, presence or absence of symptoms, and whether the hyponatremia is acute (<24 hours) or chronic (>48 hours).5
Because of Mr. W’s extremely low serum sodium concentration, predisposition to hyponatremia secondary to alcoholism, and history of severe hyponatremia, it is likely he is chronically hyponatremic.
In patients with chronic hyponatremia, neurological sequelae are associated with the need for a more rapid rate of correction of serum sodium. For most patients with chronic hyponatremia, a correction rate of ≤10 to 12 mEq/L during the first 24 hours and <18 mEq/L over 48 hours is recommended to avoid ODS.6
Evidence suggests, however, that this 1-day limit might be too high for some patients. Alcoholism, hypokalemia, malnutrition, and liver disease are present in a high percentage of patients who develop
ODS after correcting hyponatremia (Table 1).6 Therefore, for patients such as Mr. W who are at high risk of ODS, experts recommend a goal of 4 to 6 mEq/L/d with a correction rate of ≤8 mEq/L in any 24-hour period (Table 2).6
TREATMENT Sodium normalizes
Mr. W receives 1 L of normal saline in the ED before admission to the MICU. Once in the MICU, despite likely chronic hyponatremia, he receives hypertonic (3%) saline, followed by normal saline. Initially, he responds when the serum sodium concentration improves. Because of his likely SIADH, Mr. W is fluid-restricted for 4 days. Serum sodium returns to normal over 7 hospital days (Figure 2). To address the profound hypokalemia, Mr. S receives 30 mEq of potassium chloride in the ED, and potassium is repeated daily throughout his stay in the MICU.
Mr. W remains lethargic, with intermittent periods of confusion throughout the hospital stay. His altered mental status is attributed to hepatic encephalopathy secondary to alcoholic hepatitis. The Maddrey discriminant function is a calculation that stratifies patients with alcoholic hepatitis for risk of mortality and the use of steroids. Because Mr. W shows a Maddrey discriminant function ≥32, he receives methylprednisolone, followed by pentoxifylline, and liver function tests trend down. He also receives lactulose throughout hospitalization.
By discharge on hospital day 9, Mr. W’s serum sodium is 138 mEq/L; serum potassium, 4.1 mEq/L. Total bilirubin and prothrombin remain elevated. Mr. W is discharged on lactulose, thiamine, folic acid, and a 1-month course of pentoxifylline, 400 mg, 3 times a day.
READMISSION Unsteady gait, nausea
Three days after discharge, Mr. W returns to the ED after experiencing a 20-second episode of total body rigidity. He has an unsteady gait and worsening nausea and vomiting.
When Mr. W arrives in the ED, he confirms he is taking his discharge medications as prescribed. His parents report that he has consumed alcohol and Cannabis since discharge and has been taking his sibling’s prescription medications, including quetiapine.
In the ED, Mr. W is awake, alert, and oriented to person, place, and time. Vital signs are: pulse, 118 beats per minute; blood pressure, 128/73 mm Hg; respirations, 16 breaths per minute; and temperature, 98.5ºF. Physical examination, again, is notable for scleral icterus, jaundice, and asterixis. No focal neurologic deficits are noted.
Consistent with Mr. W’s previous admission, laboratory values reveal altered hepatic function and impaired coagulation. The serum sodium level remains within normal limits at 136 mEq/L. However, again, metabolic disturbances include decreased chloride (97 mEq/L), potassium (2.9 mEq/L), and CO2 (18.2 mEq/L). CT on readmission is unchanged from the earlier hospitalization.
What is your differential diagnosis for Mr. W’s total body rigidity?
a) seizure
b) ODS
c) drug intoxication
d) neuroleptic malignant syndrome
EVALUATION Shaking and weakness
Once admitted to the hospital, Mr. W reports an episode of right upper-extremity “shaking,” followed by weakness. He remembers the entire event and denies tongue biting or incontinence. He is evaluated for possible seizure, given his multiple risk factors, including drug and alcohol use, ingestion of quetiapine, and history of hyponatremia. Routine EEG is negative but prolactin level is elevated.
Mr. W’s mental status continues to wax and wane, prompting a neurology consult and MRI for further evaluation. MRI of the brain without contrast reveals restricted diffusion in the pons centrally, with extension bilaterally to the midbrain and thalami—findings consistent with central pontine myelinolysis. A neurology consultation reveals quadriparesis, paraparesis, dysarthria, and diplopia on examination, all symptoms associated with central pontine myelinolysis.
The authors’ observations
ODS, including central and extrapontine myelinolysis, is a demyelinating condition that occurs because of severe osmotic stress, most commonly secondary to the overly rapid correction of hyponatremia in patients with conditions leading to nutritional or electrolyte stress.7 Mr. W is considered at high risk of developing ODS because he fulfills the 5 criteria listed in Table 1.
Several psychiatric illnesses and neuropsychiatric medications could lead to hyponatremia. Many studies8-10 have documented hyponatremia and resulting ODS in patients with alcoholism, schizophrenia, anorexia, primary psychogenic polydipsia, and MDMA (3,4-methylenedioxymethamphetamine) abuse. Hyponatremia is a side effect of several neuropsychiatric medications, including serotonin reuptake inhibitors, lithium, tricyclic antidepressants, opioids, carbamazepine, oxcarbazepine, and antipsychotic polypharmacy. Other commonly used medications associated with hyponatremia include salt-losing diuretics, nonsteroidal anti-inflammatory drugs, and acetaminophen.7
Disease severity varies from asymptomatic to coma or death. Symptoms, although some could reverse completely, typically are a combination of neuropsychiatric (ie, emotional lability, disinhibition, and other bizarre behaviors) and neurologic. Neurologic symptoms include confusion, impaired cognition, dysarthria, dysphagia, gait instability, weakness or paralysis, and generalized seizures. Severely affected patients could experience “locked-in syndrome,” in which they are awake but unable to move or communicate. Also consistent with Mr. W’s case, ODS often presents initially with delirium, seizures, or encephalopathy, followed by a lucid interval before symptoms develop.7
Diagnosis is based on the appearance of demyelinating brain lesions on CT or MRI. MRI is more sensitive than CT; however, even an MRI scan can appear normal for as long as 4 weeks after symptoms appear.7 Therefore, an initial negative radiologic study in a high-risk patient who develops neurologic symptoms does not exclude ODS. Earlier detection is possible with diffusion-weighted MRI, which is most sensitive and can detect lesions within 24 hours of developing symptoms.11 The severity of the lesion does not correlate with severity of symptoms.
Studies reveal a considerable range in prognosis of patients with clinically symptomatic ODS. A study of 44 patients with central pontine myelinolysis, of which 42 had chronic alcoholism, reported that 34% had no significant functional deficits at follow-up, 34% had minor neurologic deficits, and 31% became dependent on personal help. Outcome did not depend on the extent or severity of neurologic symptoms or the severity of concomitant systemic complications.12
Because of its poor prognosis, prevention of ODS is important. Because ODS commonly is caused by overly rapid correction of hyponatremia, it is necessary to adhere to guidelines for treating chronic hyponatremia (Table 2). If overcorrection occurs, therapeutic re-lowering of serum sodium can be considered, but has not been validated in controlled trials. Based mainly on case reports that suggest benefit from early re-lowering serum sodium in patients with ODS symptoms, experts recommend the following:
• administer desmopressin, 2 to 4 μg, every 8 hours parenterally
• replace water orally or as 5% dextrose in water intravenously (3 mL/kg/hr)
• check serum sodium hourly until serum is reduced to goal.6
Bottom Line
Hyponatremia is the most common electrolyte disorder encountered in practice. Osmotic demyelination syndrome often is preventable, with considerable morbidity and mortality. Psychiatrists should be aware of this condition because it could be an adverse effect of many psychiatric medications and there are some psychiatric illnesses in which hyponatremia is a potential risk. In hyponatremic patients with persistent nonspecific neurologic or neuropsychiatric symptoms and negative CT imaging, additional imaging, such as MRI, is warranted.
Related Resources
- Braun MM, Barstow CH, Pyzocha NJ. Diagnosis and management of sodium disorders: hyponatremia and hypernatremia. Am Fam Physician. 2015;91(5):299-307.
- Vaidya C, Ho W, Freda BJ. Management of hyponatremia: providing treatment and avoiding harm. Cleve Clin J Med. 2010;77(10):715-726.
Drug Brand Names
Carbamazepine • Tegretol
Oxcarbazepine • Trileptal
Desmopressin • Stimate, DDAVP
Lithium • Eskalith, Lithobid
Pentoxifylline • Trental, Pentoxil
Methylprednisolone • Medrol
Quetiapine • Seroquel
Disclosure
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Elhassen EA, Schrier RW. Disorders of sodium and water balance. In: McKean SC, Ross JJ, Dressler DD, et al, eds. Principles and practice of hospital medicine. New York, NY: McGraw-Hill; 2012:2084-2093.
2. Adrogué HJ, Madias NE. Hyponatremia. N Engl J Med. 2000;342(21):1581-1589.
3. Reuler JB, Girard DE, Cooney TG. Current concepts. Wernicke’s encephalopathy. N Engl J Med. 1985;312(16):1035-1039.
4. Edelman IS, Leibman J, O’Meara MP, et al. Interrelations between serum sodium concentration, serum osmolarity and total exchangeable sodium, total exchangeable potassium and total body water. J Clin Invest. 1958;37(9):1236-1256.
5. Reynolds RM, Seckl JR. Hyponatraemia for the clinical endocrinologist. Clin Endocrinol (Oxf). 2005;63(4):366-374.
6. Verbalis JG, Goldsmith SR, Greenberg A, et al. Diagnosis, evaluation, and treatment of hyponatremia: expert panel recommendations. Am J Med. 2013;126(10 suppl 1):S1-S42.
7. Hurley RA, Filley CM, Taber KH. Central pontine myelinolysis: a metabolic disorder of myelin. J Neuropsychiatry Clin Neurosci. 2011;23(4):369-374.
8. Goldman MB. The assessment and treatment of water imbalance in patients with psychosis. Clin Schizophr Related Psychoses. 2010;4(2):115-123.
9. Patel AS, Matthews L, Bruce-Jones W. Central pontine myelinolysis as a complication of refeeding syndrome in a patient with anorexia nervosa. J Neuropsychiatry Clin Neurosci. 2008;20(3):371-373.
10. Bhuvaneswar CG, Baldessarini RJ, Harsh VL, et al. Adverse endocrine and metabolic effects of psychotropic drugs: selective clinical review. CNS Drugs. 2009;23(12):1003-1021.
11. Ruzek KA, Campeau NG, Miller GM. Early diagnosis of central pontine myelinolysis with diffusion-weighted imaging. AJNR Am J Neuroradiol. 2004;25(2):210-213.
12. Menger H, Jörg J. Outcome of central pontine and extrapontine myelinolysis (n = 44). J Neurol. 1999;246(8):700-705.
1. Elhassen EA, Schrier RW. Disorders of sodium and water balance. In: McKean SC, Ross JJ, Dressler DD, et al, eds. Principles and practice of hospital medicine. New York, NY: McGraw-Hill; 2012:2084-2093.
2. Adrogué HJ, Madias NE. Hyponatremia. N Engl J Med. 2000;342(21):1581-1589.
3. Reuler JB, Girard DE, Cooney TG. Current concepts. Wernicke’s encephalopathy. N Engl J Med. 1985;312(16):1035-1039.
4. Edelman IS, Leibman J, O’Meara MP, et al. Interrelations between serum sodium concentration, serum osmolarity and total exchangeable sodium, total exchangeable potassium and total body water. J Clin Invest. 1958;37(9):1236-1256.
5. Reynolds RM, Seckl JR. Hyponatraemia for the clinical endocrinologist. Clin Endocrinol (Oxf). 2005;63(4):366-374.
6. Verbalis JG, Goldsmith SR, Greenberg A, et al. Diagnosis, evaluation, and treatment of hyponatremia: expert panel recommendations. Am J Med. 2013;126(10 suppl 1):S1-S42.
7. Hurley RA, Filley CM, Taber KH. Central pontine myelinolysis: a metabolic disorder of myelin. J Neuropsychiatry Clin Neurosci. 2011;23(4):369-374.
8. Goldman MB. The assessment and treatment of water imbalance in patients with psychosis. Clin Schizophr Related Psychoses. 2010;4(2):115-123.
9. Patel AS, Matthews L, Bruce-Jones W. Central pontine myelinolysis as a complication of refeeding syndrome in a patient with anorexia nervosa. J Neuropsychiatry Clin Neurosci. 2008;20(3):371-373.
10. Bhuvaneswar CG, Baldessarini RJ, Harsh VL, et al. Adverse endocrine and metabolic effects of psychotropic drugs: selective clinical review. CNS Drugs. 2009;23(12):1003-1021.
11. Ruzek KA, Campeau NG, Miller GM. Early diagnosis of central pontine myelinolysis with diffusion-weighted imaging. AJNR Am J Neuroradiol. 2004;25(2):210-213.
12. Menger H, Jörg J. Outcome of central pontine and extrapontine myelinolysis (n = 44). J Neurol. 1999;246(8):700-705.
sodium concentration, Wernicke’s
encephalopathy, osmotic demyelination syndrome, electrolyte disorder
sodium concentration, Wernicke’s
encephalopathy, osmotic demyelination syndrome, electrolyte disorder
Liraglutide for obesity: New indication
Liraglutide (rDNA origin) injection, approved by the FDA in 2010 for managing type 2 diabetes mellitus (T2DM), has a new formulation and indication for treating obesity in adults as an adjunct to a reduced-calorie diet and increased physical activity (Table 1).1
Liraglutide, recommended dosage 3 mg/d (under the brand name Saxenda), is approved for adults with a body mass index (BMI) ≥30, or those with a BMI of ≥27 and a weight-related condition such as hypertension, T2DM, or high cholesterol.1 (A 1.8-mg formulation, under the brand name Victoza, is FDA-approved for managing T2DM, but is not indicated for weight management.)
How it works
Liraglutide is a glucagon-like peptide-1 (GLP-1) receptor agonist. GLP-1, which regulates appetite and calorie intake, is found in several regions of the brain that are involved in regulating appetite. Patients taking liraglutide lose weight because of decreased calorie intake, not increased energy expenditure.
Liraglutide is endogenously metabolized without a specific organ as a major route of elimination.1
Dosage and administration
Liraglutide is administered using a prefilled, multi-dose pen that can be injected in the abdomen, thigh, or upper arm. Recommended dosage is 3 mg/d, administered any time of day. Initiate dosage at 0.6 mg/d the first week, then titrate by 0.6 mg a week—to reduce the likelihood of adverse gastrointestinal symptoms—until 3 mg/d is reached.
Discontinue liraglutide if a patient has not lost at least 4% of body weight after 16 weeks of treatment, because it is unlikely the patient will achieve and sustain weight loss.
Efficacy
Liraglutide was studied in 3 clinical trials of obese and overweight participants who had a weight-related condition. Patients who had a history of major depressive disorder or suicide attempt were excluded from the studies. All participants in Studies 1 and 2 received instruction about following a reduced-calorie diet and increasing physical activity. In Study 3, patients were randomized to treatment after losing >5% of their body weight through reduced calorie intake and exercise; those who did not meet the required weight loss were excluded from the study. In these 56-week clinical studies:
• of 3,731 participants without diabetes or a weight-related comorbidity, such as high blood pressure or high cholesterol, 62% of patients (n = 2,313) who took liraglutide lost ≥5% of their body weight from baseline, compared with 34% of participants who received placebo
• of 635 participants with T2DM, 49% of patients (n = 311) treated with liraglutide lost ≥5% of their body weight compared with 16% placebo patients
• of 422 participants with a weight-related comorbidity, 42% of patients (n = 177) lost ≥5% of their body weight compared with 21.7% of placebo patients.
Improvements in some cardiovascular disease risk factors were observed. Long-term follow up was not studied.
Contraindictations
Liraglutide is contraindicated in patients who have a personal or family history of medullary thyroid carcinoma or multiple endocrine neoplasia syndrome type 2. In a 104-week study, malignant thyroid C-cell carcinomas were detected in rats and mice given liraglutide, 1 and 3 mg/kg/d; however, it was not detected in groups given 0.03 and 0.2 mg/kg/d. It isn’t known whether liraglutide can cause thyroid C-cell tumors in humans.
Patients should not take liraglutide if they have hypersensitivity to liraglutide or any product components, are using insulin, are taking any other GLP-1 receptor agonist, or are pregnant.
Adverse effects
The most common reported adverse effects are nausea (39.3%), hypoglycemia in patients with T2DM (23%), diarrhea (20.9%), constipation (19.4%), and vomiting (15.7%) (Table 2). In clinical trials, 9.8% of patients discontinued treatment because of adverse effects, compared with 4.3% of those receiving placebo.
Liraglutide has low potential for pharmacokinetic drug-drug interactions related to cytochrome P450 and plasma protein binding. For a full list of drug-drug interactions, see the full prescribing information.1
Reference
1. Saxenda [package insert]. Plainsboro, NJ: Novo Nordisk A/S; 2015.
Liraglutide (rDNA origin) injection, approved by the FDA in 2010 for managing type 2 diabetes mellitus (T2DM), has a new formulation and indication for treating obesity in adults as an adjunct to a reduced-calorie diet and increased physical activity (Table 1).1
Liraglutide, recommended dosage 3 mg/d (under the brand name Saxenda), is approved for adults with a body mass index (BMI) ≥30, or those with a BMI of ≥27 and a weight-related condition such as hypertension, T2DM, or high cholesterol.1 (A 1.8-mg formulation, under the brand name Victoza, is FDA-approved for managing T2DM, but is not indicated for weight management.)
How it works
Liraglutide is a glucagon-like peptide-1 (GLP-1) receptor agonist. GLP-1, which regulates appetite and calorie intake, is found in several regions of the brain that are involved in regulating appetite. Patients taking liraglutide lose weight because of decreased calorie intake, not increased energy expenditure.
Liraglutide is endogenously metabolized without a specific organ as a major route of elimination.1
Dosage and administration
Liraglutide is administered using a prefilled, multi-dose pen that can be injected in the abdomen, thigh, or upper arm. Recommended dosage is 3 mg/d, administered any time of day. Initiate dosage at 0.6 mg/d the first week, then titrate by 0.6 mg a week—to reduce the likelihood of adverse gastrointestinal symptoms—until 3 mg/d is reached.
Discontinue liraglutide if a patient has not lost at least 4% of body weight after 16 weeks of treatment, because it is unlikely the patient will achieve and sustain weight loss.
Efficacy
Liraglutide was studied in 3 clinical trials of obese and overweight participants who had a weight-related condition. Patients who had a history of major depressive disorder or suicide attempt were excluded from the studies. All participants in Studies 1 and 2 received instruction about following a reduced-calorie diet and increasing physical activity. In Study 3, patients were randomized to treatment after losing >5% of their body weight through reduced calorie intake and exercise; those who did not meet the required weight loss were excluded from the study. In these 56-week clinical studies:
• of 3,731 participants without diabetes or a weight-related comorbidity, such as high blood pressure or high cholesterol, 62% of patients (n = 2,313) who took liraglutide lost ≥5% of their body weight from baseline, compared with 34% of participants who received placebo
• of 635 participants with T2DM, 49% of patients (n = 311) treated with liraglutide lost ≥5% of their body weight compared with 16% placebo patients
• of 422 participants with a weight-related comorbidity, 42% of patients (n = 177) lost ≥5% of their body weight compared with 21.7% of placebo patients.
Improvements in some cardiovascular disease risk factors were observed. Long-term follow up was not studied.
Contraindictations
Liraglutide is contraindicated in patients who have a personal or family history of medullary thyroid carcinoma or multiple endocrine neoplasia syndrome type 2. In a 104-week study, malignant thyroid C-cell carcinomas were detected in rats and mice given liraglutide, 1 and 3 mg/kg/d; however, it was not detected in groups given 0.03 and 0.2 mg/kg/d. It isn’t known whether liraglutide can cause thyroid C-cell tumors in humans.
Patients should not take liraglutide if they have hypersensitivity to liraglutide or any product components, are using insulin, are taking any other GLP-1 receptor agonist, or are pregnant.
Adverse effects
The most common reported adverse effects are nausea (39.3%), hypoglycemia in patients with T2DM (23%), diarrhea (20.9%), constipation (19.4%), and vomiting (15.7%) (Table 2). In clinical trials, 9.8% of patients discontinued treatment because of adverse effects, compared with 4.3% of those receiving placebo.
Liraglutide has low potential for pharmacokinetic drug-drug interactions related to cytochrome P450 and plasma protein binding. For a full list of drug-drug interactions, see the full prescribing information.1
Liraglutide (rDNA origin) injection, approved by the FDA in 2010 for managing type 2 diabetes mellitus (T2DM), has a new formulation and indication for treating obesity in adults as an adjunct to a reduced-calorie diet and increased physical activity (Table 1).1
Liraglutide, recommended dosage 3 mg/d (under the brand name Saxenda), is approved for adults with a body mass index (BMI) ≥30, or those with a BMI of ≥27 and a weight-related condition such as hypertension, T2DM, or high cholesterol.1 (A 1.8-mg formulation, under the brand name Victoza, is FDA-approved for managing T2DM, but is not indicated for weight management.)
How it works
Liraglutide is a glucagon-like peptide-1 (GLP-1) receptor agonist. GLP-1, which regulates appetite and calorie intake, is found in several regions of the brain that are involved in regulating appetite. Patients taking liraglutide lose weight because of decreased calorie intake, not increased energy expenditure.
Liraglutide is endogenously metabolized without a specific organ as a major route of elimination.1
Dosage and administration
Liraglutide is administered using a prefilled, multi-dose pen that can be injected in the abdomen, thigh, or upper arm. Recommended dosage is 3 mg/d, administered any time of day. Initiate dosage at 0.6 mg/d the first week, then titrate by 0.6 mg a week—to reduce the likelihood of adverse gastrointestinal symptoms—until 3 mg/d is reached.
Discontinue liraglutide if a patient has not lost at least 4% of body weight after 16 weeks of treatment, because it is unlikely the patient will achieve and sustain weight loss.
Efficacy
Liraglutide was studied in 3 clinical trials of obese and overweight participants who had a weight-related condition. Patients who had a history of major depressive disorder or suicide attempt were excluded from the studies. All participants in Studies 1 and 2 received instruction about following a reduced-calorie diet and increasing physical activity. In Study 3, patients were randomized to treatment after losing >5% of their body weight through reduced calorie intake and exercise; those who did not meet the required weight loss were excluded from the study. In these 56-week clinical studies:
• of 3,731 participants without diabetes or a weight-related comorbidity, such as high blood pressure or high cholesterol, 62% of patients (n = 2,313) who took liraglutide lost ≥5% of their body weight from baseline, compared with 34% of participants who received placebo
• of 635 participants with T2DM, 49% of patients (n = 311) treated with liraglutide lost ≥5% of their body weight compared with 16% placebo patients
• of 422 participants with a weight-related comorbidity, 42% of patients (n = 177) lost ≥5% of their body weight compared with 21.7% of placebo patients.
Improvements in some cardiovascular disease risk factors were observed. Long-term follow up was not studied.
Contraindictations
Liraglutide is contraindicated in patients who have a personal or family history of medullary thyroid carcinoma or multiple endocrine neoplasia syndrome type 2. In a 104-week study, malignant thyroid C-cell carcinomas were detected in rats and mice given liraglutide, 1 and 3 mg/kg/d; however, it was not detected in groups given 0.03 and 0.2 mg/kg/d. It isn’t known whether liraglutide can cause thyroid C-cell tumors in humans.
Patients should not take liraglutide if they have hypersensitivity to liraglutide or any product components, are using insulin, are taking any other GLP-1 receptor agonist, or are pregnant.
Adverse effects
The most common reported adverse effects are nausea (39.3%), hypoglycemia in patients with T2DM (23%), diarrhea (20.9%), constipation (19.4%), and vomiting (15.7%) (Table 2). In clinical trials, 9.8% of patients discontinued treatment because of adverse effects, compared with 4.3% of those receiving placebo.
Liraglutide has low potential for pharmacokinetic drug-drug interactions related to cytochrome P450 and plasma protein binding. For a full list of drug-drug interactions, see the full prescribing information.1
Reference
1. Saxenda [package insert]. Plainsboro, NJ: Novo Nordisk A/S; 2015.
Reference
1. Saxenda [package insert]. Plainsboro, NJ: Novo Nordisk A/S; 2015.
Managing first-episode psychosis: An early stage of schizophrenia with distinct treatment needs
The less time that passes between the onset of psychosis and initiation of appropriate treatment, the greater the patient’s odds of recovery.1 However, relapse prevention is a major clinical challenge because >80% of patients will relapse within 5 years, and, on average, 40% to 50% of patients with a first-episode schizophrenia will relapse within 2 years depending on the definition used and patient characteristics.2 Although there are several explanations and contributing factors to relapses, nonadherence—partial or complete discontinuation of antipsychotics—is a primary risk factor, contributing to a 5-fold increase in relapse risk.3
As such, optimal antipsychotic selection, dosing, and monitoring play an important role in managing this illness. Patients with first-episode psychosis (FEP) are unusual in some ways, compared with patients with multiple episodes of psychosis and represent a different stage of schizophrenia.
In this 2-part series, we will discuss pharmacotherapy for FEP. This article focuses on antipsychotic selection, dosage, and duration of treatment among these patients. The second article, in the July 2015 issue, reviews the rationale and evidence for non-standard, first-line therapies, including long-acting injectable antipsychotics and clozapine.
Defining FEP
FEP refers to a patient who has presented, been evaluated, and received treatment for the first psychotic episode associated with a schizophrenia spectrum diagnosis.4 FEP is part of a trajectory marked by tran sitional periods. The patient transitions from being “healthy” to a prodromal state characterized by: (1) nonpsychotic behavioral disturbances such as depression or obsessive-compulsive disorder, (2) attenuated psychotic symptoms not requiring treatment, then converting to (3) psychotic symptoms prompting initial presentation for antipsychotic pharmacotherapy, leading to (4) a formal diagnosis of schizophreniform disorder and, subsequently, schizophrenia, requiring treatment to stabilize symptoms.
There are 2 critical periods along this continuum: prodromal stage and the duration of untreated psychosis (DUP). The prodromal period is a retrospectively identified time where the patient shows initial nonpsychotic disturbances (eg, cognitive and behavioral symptoms) before exhibiting clinical diagnostic criteria for a schizophrenia spectrum disorder. Approximately one-third of patients exhibiting these symptoms convert to psychosis within 1 year, and early treatment engagement at this stage has been shown to improve outcomes.5 The DUP is the time from when a patient has noticeable psychotic symptoms to initiation of drug treatment. The DUP is a consistent predictor of clinical outcome in schizophrenia, including negative symptoms, quality of life, and functional capacity.1
Antipsychotic selection
Treatment goals for FEP patients include:
• minimizing the DUP
• rapidly stabilizing psychosis
• achieving full symptomatic remission
• preventing relapse.
Several treatment guidelines for managing schizophrenia offer variable recommendations for initial antipsychotic treatment in patients with first-episode schizophrenia (Table 1).6-15 Most recommend second-generation antipsychotics (SGAs) over first-generation antipsychotics (FGAs)6,8,9,13,15 with specific recommendations on minimizing neurologic and metabolic adverse effects—to which FEP patients are susceptible—by avoiding high-potency and neurotoxic FGAs (eg, haloperidol and fluphenazine),7 clozapine,11,14 olanzapine,11 or ziprasidone.14 Two guidelines—the National Institute for Health and Care Excellence and the Scottish Intercollegiate Guidelines Network—do not state a preference for antipsychotic selection.10,12
The rationale for these recommendations is based on efficacy data, tolerability differences, FDA-approved indications, and recent FDA approvals with sparse post-marketing data. Of note, there are a lack of robust data for newer antipsychotics (eg, aripiprazole, paliperidone, iloperidone, asenapine, and lurasidone) in effectively and safely treating FEP; however, given the results of other antipsychotics studies, it is likely the efficacy and tolerability of these drugs can be extrapolated from experience with multi-episode patients.
Study design and demographics. Research studies of FEP share some similarities in study design; however, there is enough variability to make it difficult to compare studies and generalize findings (Table 2).16 The variability of DUP is a limitation when comparing studies because it is a significant predictor of clinical outcome. Patients who abuse substances—and often are more challenging to treat17—typically are excluded from these trials, which could explain the high response rate documented in studies of first-episode schizophrenia.
In addition, some FEP patients included in clinical trials might not be truly antipsychotic naïve; an estimated 25% to 75% of patients in these studies are antipsychotic naïve. This is an important consideration when comparing data on adverse effects that occur early in treatment. Additionally, acknowledging the advantages and disadvantages of how to handle missing data is critical because of the high dropout rate observed in these studies.18
Efficacy. There is a high response rate to antipsychotic therapy—ranging from 46% to 96%, depending on the study—in patients with first-episode schizophrenia.3 The response mainly is seen in reduction of positive symptoms because typically negative and cognitive symptoms do not respond to antipsychotics. One study reported only 29% of patients achieved both positive and negative symptom remission.19 It is likely that secondary negative symptoms caused by social withdrawal, reduced speech, and avoidance improve when positive symptoms subside, but primary negative symptoms endure.In general, there is a lack of evidence suggesting that 1 antipsychotic class or agent is more effective than another. Studies mainly assess effectiveness using the primary outcome measure of all-cause discontinuation, such as the Clinical Antipsychotic Trials of Intervention Effectiveness study.20 This outcome measure is a mixture of patient preference, tolerability, and efficacy that provides a more generalizable gauge on how well the treatment works in the clinic rather than tightly regulated settings such as clinical trials. A recent meta-analysis supports no differences in efficacy among antipsychotics in early-episode psychosis.21
Tolerability. Because there are no significant differences among antipsychotic classes or agents in terms of efficacy in first-episode schizophrenia, drug selection is guided mainly by (1) the adverse effect profile and (2) what should be avoided depending on patient-specific variables. Evidence suggests first-episode patients are more sensitive to adverse effects of antipsychotics, particularly neurologic side effects (see this article at CurrentPsychiatry.com for a table comparing adverse effects of antipsychotics in first-episode psychosis).18,22-29 Overall adverse effect profiles remain similar across FEP or multi-episode patients, but tend to be more exaggerated in drug-naïve patients with FEP.
Regarding FGA side effects, McEvoy et al18 demonstrated the neuroleptic threshold occurs at 50% lower haloperidol dosages in patients with first-episode schizophrenia (2.1 mg/d) compared with multi-episode schizophrenia (4.3 mg/d). Other trials suggest SGAs are associated with a lower risk of extrapyramidal side effects (EPS) or use of adjunctive therapies such as anticholinergic drugs or benzodiazepines.23-27 An exception to this statement is that higher risperidone dosages (≥4 to 6 mg/d) have been found to have higher rates of EPS and use of adjunctive medications to treat these symptoms in FEP.26 This is important because studies report higher discontinuation rates with more severe adverse effects of antipsychotics.
Cardiometabolic effects are of particular concern in first-episode patients because most weight gain happens in the first 3 to 4 months of treatment and remains throughout the first year.18,24,29,30 Studies have shown that olanzapine, quetiapine, and risperidone are associated with more clinically significant weight gain compared with haloperidol and ziprasidone.23-25 Olanzapine-associated weight gain has been reported to be twice that of quetiapine and risperidone.18 Regardless, the EUFEST trial did not find a difference in clinically significant weight gain after 12 months among the antipsychotics studied, including haloperidol and ziprasidone.25
Weight gain associated with these antipsychotics is accompanied by changes in fasting triglycerides, glucose, total cholesterol,23 and high-density lipoprotein cholesterol as well as an increase in body mass index (BMI) categorization29 (eg, shift from normal to overweight).18,25 Patients with lower baseline BMI and in racial minority groups might experience more rapid weight gain regardless of antipsychotic selection.29,30
Hyperprolactinemia could be under-recognized and could contribute to early treatment discontinuation.31 Evidence in patients with first-episode schizophrenia suggests similar outcomes as those seen in multi-episode patients, in whom risperidone is associated with higher prolactin elevations and clinically significant hyperprolactinemia (eg, galactorrhea and gynecomastia) compared with olanzapine, quetiapine, and low-dose haloperidol.18,23,24 However, there is a lack of studies that assess whether long-term therapy with strong D2 receptor antagonists increases the risk of bone demineralization or pathological fractures when started before patients’ bones reach maximum density in their mid-20s.31
Antipsychotic dosing
Given the high rate of treatment response in FEP and patients’ higher sensitivity to antipsychotic adverse effects, particularly EPS, guidelines recommend antipsychotic dosages lower than those used for multi-episode schizophrenia,11 especially FGAs. Based on trial data, commonly used dosages include:
• haloperidol, ≤5 mg/d23-25,29
• olanzapine, 10 mg/d18,23,25,29
• risperidone, ≤4 to 6 mg/d.18,24,29,32
In general, haloperidol and risperidone, 2 to 3 mg/d, were well tolerated and effective in trials. Higher quetiapine dosages of 500 to 600 mg/d could be required.11,18,25
According to a survey on prescribing practices of antipsychotic selection and dosing in first-episode schizophrenia,4 clinical prescribing practices tend to use unnecessarily high initial antipsychotic dosing compared with trial data. There also is variability in the usual target antipsychotic dosage ranging from 50% lower dosages to normal dosages in chronic schizophrenia to above FDA-approved maximum dosages for olanzapine (which may be necessary to counteract tobacco-induced cytochrome P450 1A2 enzyme induction).
In addition, these clinicians reported prescribing aripiprazole, an antipsychotic with weaker evidence (eg, case reports, case series, open-label studies) supporting its efficacy and tolerability in FEP. These prescribing practices could reflect attempts to reduce the DUP and achieve symptom remission, so long as tolerability is not a concern.
Essentially, prescribed dosages should be based on symptom improvement and tolerability. This ideal dosage will vary as illustrated by Kapur et al,33 who reported that FEP patients (N = 20) given haloperidol, 1 mg or 2.5 mg/d, had D2 receptor occupancy rates of 38% to 87%, which was significantly dose-related (1 mg/d mean = 59%, 2.5 mg/d mean = 75%). Clinical response and EPS significantly increased as D2 receptor occupancy exceeded 65% and 78%, respectively.
Antipsychotic response
When should you expect to see symptom improvement in patients with first-episode schizophrenia?
Emsley et al34 reported a 77.6% response rate among first-episode patients (N = 522) treated with low dosages of risperidone (mean modal dosage [MMD] = 3.3 mg/d) and haloperidol (MMD = 2.9 mg/d). They found variable response times that were evenly dispersed over a 10-week period. Nearly one-quarter (22.5%) did not respond until after week 4 and 11.2% did not respond until after week 8. In a study of FEP patients (N = 112) treated with olanzapine (MMD = 11.8 mg/d) or risperidone (MMD = 3.9 mg/d), Gallego et al35 reported a cumulative response of 39.6% at week 8 and 65.1% at week 16.
Although there is evidence that, among multi-episode patients, early nonresponse to antipsychotic therapy could predict subsequent nonresponse,36 the evidence is mixed for first-episode schizophrenia. Studies by Emsley et al34 and Gallego et al35 did not find that early nonresponse at weeks 1 or 2 predicted subsequent nonresponse at week 4 or later. However, other studies support the idea that early nonresponse predicts subsequent nonresponse and early antipsychotic response predicts future response in first-episode patients, with good specificity and sensitivity.37,38
Overall, treatment response in first-episode schizophrenia is variable. An adequate antipsychotic trial may be longer, 8 to 16 weeks, compared with 4 to 8 weeks in multi-episode patients. Because research suggests that failure to respond to treatment may lead to medication nonadherence,39 it is reasonable to consider switching antipsychotics when a patient experiences minimal or no response to antipsychotic therapy at week 2; however, this should be a patient-specific decision.
How long should you continue therapy after symptom remission?
There is a lack of consensus on the duration of therapy for a patient treated for first-episode schizophrenia because a small percentage (10% to 20%) do not relapse after the first psychotic episode.3 In general, treatment guidelines and expert consensus statements recommend at least 1 to 2 years of treatment before considering a discontinuation trial.7,10-11 Discuss the benefits and risks of maintenance treatment with your patient and obtain informed consent. With patients with minimal insight, obtaining proper consent is not possible and the physician must exercise judgment unilaterally, if necessary, after educating the family.
After at least 12 months of treatment, antipsychotic therapy could continue indefinitely, depending on patient-specific factors. There are no predictors for identifying patients who do not require maintenance therapy beyond the first psychotic episode. The absence of negative and cognitive deficits could provide clues that a patient might be a candidate for antipsychotic tapering.
Predicting the treatment course
Research investigating clinical predictors or biomarkers that forecast whether a patient will respond to treatment is preliminary. Many characteristics have been identified (Table 31,3,4,23,25,40) and include shorter DUP,1 poorer premorbid function,3 antipsychotic discontinuation,3 a trusting patient-doctor relationship,41 and antipsychotic-related adverse effects,23,25 which are predictive of response, nonresponse, relapse, adherence, and nonadherence, respectively.
Bottom Line
The goals of pharmacological treatment of first-episode schizophrenia are to minimize the duration of untreated psychosis and target full remission of positive symptoms using the lowest possible antipsychotic dosages. Pharmacotherapy should continued for 1 to 2 years, with longer duration considered if it is discussed with the patient and with vigilant monitoring for adverse effects and suboptimal medication nonadherence to prevent relapse.
Editor’s note: The second article in this series in the July 2015 issue reviews the rationale and evidence for non-standard, first-line therapies, including long-acting injectable antipsychotics and clozapine.
Related Resources
• Recovery After an Initial Schizophrenia Episode (RAISE) Project Early Treatment Program. National Institute of Mental Health. http://raiseetp.org.
• Martens L, Baker S. Promoting recovery from first episode psychosis: a guide for families. Centre for Addiction and Mental Health. http://www.camh.ca/en/hospital/ Documents/www.camh.net/AboutCAMH/Guideto CAMH/MentalHealthPrograms/SchizophreniaProgram/ 3936PromotingRecoveryFirstEpisodePsychosisfinal.pdf.
Drug Brand Names
Aripiprazole • Abilify Lurasidone • Latuda
Asenapine • Saphris Olanzapine • Zyprexa
Clozapine • Clozaril Paliperidone • Invega
Fluphenazine • Prolixin Quetiapine • Seroquel
Iloperidone • Fanapt Risperidone • Risperdal
Haloperidol • Haldol Ziprasidone • Geodon
Disclosures
Dr. Gardner reports no financial relationships with any companies whose products are mentioned in this article or with manufacturers of competing products.
Dr. Nasrallah is a consultant to Acadia, Alkermes, Lundbeck, Janssen, Merck, Otsuka, and Sunovion, and is a speaker for Alkermes, Lundbeck, Janssen, Otsuka, and Sunovion.
1. Perkins DO, Gu H, Boteva K, et al. Relationship between duration of untreated psychosis and outcome in first-episode schizophrenia: a critical review and meta-analysis. Am J Psychiatry. 2005;162(10):1785-1804.
2. Bradford DW, Perkins DO, Lieberman JA. Pharmacological management of first-episode schizophrenia and related nonaffective psychoses. Drugs. 2003;63(21):2265-2283.
3. Robinson D, Woerner MG, Alvir JM, et al. Predictors of relapse following a response from a first episode of schizophrenia or schizoaffective disorder. Arch Gen Psychiatry. 1999;56(3):241-247.
4. Weiden PJ, Buckley PF, Grody M. Understanding and treating “first-episode” schizophrenia. Psychiatr Clin North Am. 2007;30(3):481-510.
5. Madaan V, Bestha DP, Kolli V. Schizophrenia prodrome: an optimal approach. Current Psychiatry. 2014;13(3):16-20, 29-30.
6. Lehman AF, Lieberman JA, Dixon LB, et al; American Psychiatric Association; Steering Committee on Practice Guidelines. Practice guideline for the treatment of patients with schizophrenia, second edition. Am J Psychiatry. 2004;161(suppl 2):1-56.
7. Barnes TR; Schizophrenia Consensus Group of British Association for Psychopharmacology. Evidence-based guidelines for the pharmacological treatment of schizophrenia: recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2011;25(5):567-620.
8. Canadian Psychiatric Association. Clinical practice guideline. Treatment of schizophrenia. Can J Psychiatry. 2005;50(13 suppl 1):7S-57S.
9. McEvoy JP, Scheifler PL, Frances A. Treatment of schizophrenia 1999. Expert consensus guideline series. J Clin Psychiatry. 1999;60(suppl 11):4-80.
10. National Institute for Health and Care Excellence (NICE). Clinical guideline 178: Psychosis and schizophrenia in adults: treatment and management. London, United Kingdom: National Institute for Health and Care Excellence (NICE); 2014.
11. Buchanan RW, Kreyenbuhl J, Kelly DL, et al; Schizophrenia Patient Outcomes Research Team (PORT). The 2009 schizophrenia PORT psychopharmacological treatment recommendations and summary statements. Schizophr Bull. 2010;36(1):71-93.
12. Scottish Intercollegiate Guidelines Network (SIGN). Management of schizophrenia. Edinburgh, Scotland: Scottish Intercollegiate Guidelines Network; 2013. SIGN publication no. 131.
13. Argo TR, Crismon ML, Miller AL, et al. Texas Medication Algorithm Project procedural manual. Schizophrenia treatment algorithms. Austin, Texas: Texas Department of State Health Services; 2008.
14. Marder SR, Essock SM, Miller Al, et al. The Mount Sinai conference on the pharmacotherapy of schizophrenia. Schizophr Bull. 2002;28(1):5-16.
15. Bandelow B, Zohar J, Hollander E, et al; WFSBP Task Force on Treatment Guidelines for Anxiety, Obsessive-Compulsive and Post-Traumatic Stress Disorders. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry. 2008;9(4):248-312.
16. Robinson DG, Woerner MG, Alvir JMJ, et al. Predictors of relapse following response from a first episode of schizophrenia or schizoaffective disorder. Arch Gen Psych. 1999;56(3):241-247.
17. Green AI, Tohen MF, Hamer RM, et al. First episode schizophrenia-related psychosis and substance use disorders: acute response to olanzapine and haloperidol. Schizophr Res. 2004;66(2-3):125-135.
18. McEvoy JP, Lieberman JA, Perkins DO, et al. Efficacy and tolerability of olanzapine, quetiapine, and risperidone in the treatment of early psychosis: a randomized, double-blind 52-week comparison. Am J Psychiatry. 2007;164(7): 1050-1060.
19. Henry LP, Amminger GP, Harris MG, et al. The EPPIC follow-up study of first-episode psychosis: longer-term clinical and functional outcome 7 years after index admission. J Clin Psychiatry. 2010;71(6):716-728.
20. Lieberman JA, Stroup TS, McEvoy JP, et al; Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Investigators. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. New Engl J Med. 2005; 353(12):1209-1223.
21. Crossley NA, Constante M, McGuire P, et al. Efficacy of atypical v. typical antipsychotics in the treatment of early psychosis: meta-analysis. Br J Psychiatry. 2010;196(6):434-439.
22. McEvoy JP, Hogarty GE, Steingard S. Optimal dose of neuroleptic in acute schizophrenia: a controlled study of the neuroleptic threshold and higher haloperidol dose. Arch Gen Psych. 1991;48(8):739-745.
23. Lieberman JA, Tollefson G, Tohen M, et al; HGDH Study Group. Comparative efficacy and safety of atypical and conventional antipsychotic drugs in first-episode psychosis: a randomized, double-blind trial of olanzapine versus haloperidol. Am J Psychiatry. 2003;160(8):1396-1404.
24. Schooler N, Rabinowitz J, Davidson M, et al; Early Psychosis Global Working Group. Risperidone and haloperidol in first-episode psychosis: a long-term randomized trial. Am J Psychiatry. 2005;162(5):947-953.
25. Kahn RS, Fleischhacker WW, Boter H, et al; EUFEST study group. Effectiveness of antipsychotic drugs in first-episode schizophrenia and schizophreniform disorder: an open randomised clinical trial. Lancet. 2008;371(9618):1085-1097.
26. Emsley RA; Risperidone Working Group. Risperidone in the treatment of first-episode psychotic patients: a double-blind multicenter study. Schizophr Bull. 1999;25(4):721-729.
27. Lieberman JA, Phillips M, Gu H, et al. Atypical and conventional antipsychotic drugs in treatment-naïve first-episode schizophrenia: a 52-week randomized trial of clozapine vs chlorpromazine. Neuropsychopharmacology. 2003;28(5):995-1003.
28. Girgis RR, Phillips MR, Li X, et al. Clozapine v. chlorpromazine in treatment-naive, first-episode schizophrenia: 9-year outcomes of a randomised clinical trial. Br J Psychiatry. 2011;199(4):281-288.
29. Robinson DG, Woerner MG, Napolitano B, et al. Randomized comparison of olanzapine versus risperidone for the treatment of first-episode schizophrenia: 4-month outcomes. Am J Psychiatry. 2006;163(12):2096-2102.
30. Zipursky RB, Gu H, Green AI, et al. Course and predictors of weight gain in people with first-episode psychosis treated with olanzapine or haloperidol. Br J Psychiatry. 2005;187:537-543.
31. Taylor M, Waight A, Leonard B. Advances in the understanding and challenges facing the management of first-episode schizophrenia. J Psychopharmacol. 2012; 26(suppl 5):3-5.
32. Merlo MC, Hofer H, Gekle W, et al. Risperidone, 2mg/day vs. 4mg/day, in first-episode, acutely psychotic patients: treatment efficacy and effects on fine motor functioning. J Clin Psychiatry. 2002;63(10):885-891.
33. Kapur S, Zipursky R, Jones C, et al. Relationship between dopamine D2 occupancy, clinical response, and side effects: a double-blind PET study of first-episode schizophrenia. Am J Psychiatry. 2000;157(4):514-520.
34. Emsley R, Rabinowitz J, Medori R. Time course for antipsychotic treatment response in first-episode schizophrenia. Am J Psychiatry. 2006;163(4):743-745.
35. Gallego JA, Robinson DG, Sevy SM, et al. Time to treatment response in first-episode schizophrenia: should acute treatment trials last several months? J Clin Psychiatry. 2011;72(12):1691-1696.
36. Gardner KN, Bostwick JR. Antipsychotic treatment response in schizophrenia. Am J Health Sys Pharm. 2012;69(21):1872-1879.
37. Stauffer VL, Case M, Kinon BJ, et al. Early response to antipsychotic therapy as a clinical marker of subsequent response in the treatment of patients with first-episode psychosis. Psychiatry Res. 2011;187(1-2):42-48.
38. Schennach-Wolff R, Seemüller FH, Mayr A, et al. An early improvement threshold to predict response and remission in first-episode schizophrenia. Br J Psychiatry. 2010;196(6):460-466.
39. Perkins DO, Gu H, Weiden PJ, et al; Comparison of Atypicals in First Episode study group. Predictors of treatment discontinuation and medication nonadherence in patients recovering from a first episode of schizophrenia, schizophreniform disorder, or schizoaffective disorder: a randomized, double-blind, flexible-dose, multicenter study. J Clin Psychiatry. 2008;69(1):106-113.
40. Garner B, Berger GE, Nicolo JP, et al. Pituitary volume and early treatment response in drug-naïve first-episode psychosis patients. Schizophr Res. 2009;113(1):65-71.
41. Sapra M, Weiden PJ, Schooler NR, et al. Reasons for adherence and nonadherence: a pilot study comparing first-and multi-episode schizophrenia patients. Clin Schizophr Relat Psychoses. 2014;7(4):199-206.
The less time that passes between the onset of psychosis and initiation of appropriate treatment, the greater the patient’s odds of recovery.1 However, relapse prevention is a major clinical challenge because >80% of patients will relapse within 5 years, and, on average, 40% to 50% of patients with a first-episode schizophrenia will relapse within 2 years depending on the definition used and patient characteristics.2 Although there are several explanations and contributing factors to relapses, nonadherence—partial or complete discontinuation of antipsychotics—is a primary risk factor, contributing to a 5-fold increase in relapse risk.3
As such, optimal antipsychotic selection, dosing, and monitoring play an important role in managing this illness. Patients with first-episode psychosis (FEP) are unusual in some ways, compared with patients with multiple episodes of psychosis and represent a different stage of schizophrenia.
In this 2-part series, we will discuss pharmacotherapy for FEP. This article focuses on antipsychotic selection, dosage, and duration of treatment among these patients. The second article, in the July 2015 issue, reviews the rationale and evidence for non-standard, first-line therapies, including long-acting injectable antipsychotics and clozapine.
Defining FEP
FEP refers to a patient who has presented, been evaluated, and received treatment for the first psychotic episode associated with a schizophrenia spectrum diagnosis.4 FEP is part of a trajectory marked by tran sitional periods. The patient transitions from being “healthy” to a prodromal state characterized by: (1) nonpsychotic behavioral disturbances such as depression or obsessive-compulsive disorder, (2) attenuated psychotic symptoms not requiring treatment, then converting to (3) psychotic symptoms prompting initial presentation for antipsychotic pharmacotherapy, leading to (4) a formal diagnosis of schizophreniform disorder and, subsequently, schizophrenia, requiring treatment to stabilize symptoms.
There are 2 critical periods along this continuum: prodromal stage and the duration of untreated psychosis (DUP). The prodromal period is a retrospectively identified time where the patient shows initial nonpsychotic disturbances (eg, cognitive and behavioral symptoms) before exhibiting clinical diagnostic criteria for a schizophrenia spectrum disorder. Approximately one-third of patients exhibiting these symptoms convert to psychosis within 1 year, and early treatment engagement at this stage has been shown to improve outcomes.5 The DUP is the time from when a patient has noticeable psychotic symptoms to initiation of drug treatment. The DUP is a consistent predictor of clinical outcome in schizophrenia, including negative symptoms, quality of life, and functional capacity.1
Antipsychotic selection
Treatment goals for FEP patients include:
• minimizing the DUP
• rapidly stabilizing psychosis
• achieving full symptomatic remission
• preventing relapse.
Several treatment guidelines for managing schizophrenia offer variable recommendations for initial antipsychotic treatment in patients with first-episode schizophrenia (Table 1).6-15 Most recommend second-generation antipsychotics (SGAs) over first-generation antipsychotics (FGAs)6,8,9,13,15 with specific recommendations on minimizing neurologic and metabolic adverse effects—to which FEP patients are susceptible—by avoiding high-potency and neurotoxic FGAs (eg, haloperidol and fluphenazine),7 clozapine,11,14 olanzapine,11 or ziprasidone.14 Two guidelines—the National Institute for Health and Care Excellence and the Scottish Intercollegiate Guidelines Network—do not state a preference for antipsychotic selection.10,12
The rationale for these recommendations is based on efficacy data, tolerability differences, FDA-approved indications, and recent FDA approvals with sparse post-marketing data. Of note, there are a lack of robust data for newer antipsychotics (eg, aripiprazole, paliperidone, iloperidone, asenapine, and lurasidone) in effectively and safely treating FEP; however, given the results of other antipsychotics studies, it is likely the efficacy and tolerability of these drugs can be extrapolated from experience with multi-episode patients.
Study design and demographics. Research studies of FEP share some similarities in study design; however, there is enough variability to make it difficult to compare studies and generalize findings (Table 2).16 The variability of DUP is a limitation when comparing studies because it is a significant predictor of clinical outcome. Patients who abuse substances—and often are more challenging to treat17—typically are excluded from these trials, which could explain the high response rate documented in studies of first-episode schizophrenia.
In addition, some FEP patients included in clinical trials might not be truly antipsychotic naïve; an estimated 25% to 75% of patients in these studies are antipsychotic naïve. This is an important consideration when comparing data on adverse effects that occur early in treatment. Additionally, acknowledging the advantages and disadvantages of how to handle missing data is critical because of the high dropout rate observed in these studies.18
Efficacy. There is a high response rate to antipsychotic therapy—ranging from 46% to 96%, depending on the study—in patients with first-episode schizophrenia.3 The response mainly is seen in reduction of positive symptoms because typically negative and cognitive symptoms do not respond to antipsychotics. One study reported only 29% of patients achieved both positive and negative symptom remission.19 It is likely that secondary negative symptoms caused by social withdrawal, reduced speech, and avoidance improve when positive symptoms subside, but primary negative symptoms endure.In general, there is a lack of evidence suggesting that 1 antipsychotic class or agent is more effective than another. Studies mainly assess effectiveness using the primary outcome measure of all-cause discontinuation, such as the Clinical Antipsychotic Trials of Intervention Effectiveness study.20 This outcome measure is a mixture of patient preference, tolerability, and efficacy that provides a more generalizable gauge on how well the treatment works in the clinic rather than tightly regulated settings such as clinical trials. A recent meta-analysis supports no differences in efficacy among antipsychotics in early-episode psychosis.21
Tolerability. Because there are no significant differences among antipsychotic classes or agents in terms of efficacy in first-episode schizophrenia, drug selection is guided mainly by (1) the adverse effect profile and (2) what should be avoided depending on patient-specific variables. Evidence suggests first-episode patients are more sensitive to adverse effects of antipsychotics, particularly neurologic side effects (see this article at CurrentPsychiatry.com for a table comparing adverse effects of antipsychotics in first-episode psychosis).18,22-29 Overall adverse effect profiles remain similar across FEP or multi-episode patients, but tend to be more exaggerated in drug-naïve patients with FEP.
Regarding FGA side effects, McEvoy et al18 demonstrated the neuroleptic threshold occurs at 50% lower haloperidol dosages in patients with first-episode schizophrenia (2.1 mg/d) compared with multi-episode schizophrenia (4.3 mg/d). Other trials suggest SGAs are associated with a lower risk of extrapyramidal side effects (EPS) or use of adjunctive therapies such as anticholinergic drugs or benzodiazepines.23-27 An exception to this statement is that higher risperidone dosages (≥4 to 6 mg/d) have been found to have higher rates of EPS and use of adjunctive medications to treat these symptoms in FEP.26 This is important because studies report higher discontinuation rates with more severe adverse effects of antipsychotics.
Cardiometabolic effects are of particular concern in first-episode patients because most weight gain happens in the first 3 to 4 months of treatment and remains throughout the first year.18,24,29,30 Studies have shown that olanzapine, quetiapine, and risperidone are associated with more clinically significant weight gain compared with haloperidol and ziprasidone.23-25 Olanzapine-associated weight gain has been reported to be twice that of quetiapine and risperidone.18 Regardless, the EUFEST trial did not find a difference in clinically significant weight gain after 12 months among the antipsychotics studied, including haloperidol and ziprasidone.25
Weight gain associated with these antipsychotics is accompanied by changes in fasting triglycerides, glucose, total cholesterol,23 and high-density lipoprotein cholesterol as well as an increase in body mass index (BMI) categorization29 (eg, shift from normal to overweight).18,25 Patients with lower baseline BMI and in racial minority groups might experience more rapid weight gain regardless of antipsychotic selection.29,30
Hyperprolactinemia could be under-recognized and could contribute to early treatment discontinuation.31 Evidence in patients with first-episode schizophrenia suggests similar outcomes as those seen in multi-episode patients, in whom risperidone is associated with higher prolactin elevations and clinically significant hyperprolactinemia (eg, galactorrhea and gynecomastia) compared with olanzapine, quetiapine, and low-dose haloperidol.18,23,24 However, there is a lack of studies that assess whether long-term therapy with strong D2 receptor antagonists increases the risk of bone demineralization or pathological fractures when started before patients’ bones reach maximum density in their mid-20s.31
Antipsychotic dosing
Given the high rate of treatment response in FEP and patients’ higher sensitivity to antipsychotic adverse effects, particularly EPS, guidelines recommend antipsychotic dosages lower than those used for multi-episode schizophrenia,11 especially FGAs. Based on trial data, commonly used dosages include:
• haloperidol, ≤5 mg/d23-25,29
• olanzapine, 10 mg/d18,23,25,29
• risperidone, ≤4 to 6 mg/d.18,24,29,32
In general, haloperidol and risperidone, 2 to 3 mg/d, were well tolerated and effective in trials. Higher quetiapine dosages of 500 to 600 mg/d could be required.11,18,25
According to a survey on prescribing practices of antipsychotic selection and dosing in first-episode schizophrenia,4 clinical prescribing practices tend to use unnecessarily high initial antipsychotic dosing compared with trial data. There also is variability in the usual target antipsychotic dosage ranging from 50% lower dosages to normal dosages in chronic schizophrenia to above FDA-approved maximum dosages for olanzapine (which may be necessary to counteract tobacco-induced cytochrome P450 1A2 enzyme induction).
In addition, these clinicians reported prescribing aripiprazole, an antipsychotic with weaker evidence (eg, case reports, case series, open-label studies) supporting its efficacy and tolerability in FEP. These prescribing practices could reflect attempts to reduce the DUP and achieve symptom remission, so long as tolerability is not a concern.
Essentially, prescribed dosages should be based on symptom improvement and tolerability. This ideal dosage will vary as illustrated by Kapur et al,33 who reported that FEP patients (N = 20) given haloperidol, 1 mg or 2.5 mg/d, had D2 receptor occupancy rates of 38% to 87%, which was significantly dose-related (1 mg/d mean = 59%, 2.5 mg/d mean = 75%). Clinical response and EPS significantly increased as D2 receptor occupancy exceeded 65% and 78%, respectively.
Antipsychotic response
When should you expect to see symptom improvement in patients with first-episode schizophrenia?
Emsley et al34 reported a 77.6% response rate among first-episode patients (N = 522) treated with low dosages of risperidone (mean modal dosage [MMD] = 3.3 mg/d) and haloperidol (MMD = 2.9 mg/d). They found variable response times that were evenly dispersed over a 10-week period. Nearly one-quarter (22.5%) did not respond until after week 4 and 11.2% did not respond until after week 8. In a study of FEP patients (N = 112) treated with olanzapine (MMD = 11.8 mg/d) or risperidone (MMD = 3.9 mg/d), Gallego et al35 reported a cumulative response of 39.6% at week 8 and 65.1% at week 16.
Although there is evidence that, among multi-episode patients, early nonresponse to antipsychotic therapy could predict subsequent nonresponse,36 the evidence is mixed for first-episode schizophrenia. Studies by Emsley et al34 and Gallego et al35 did not find that early nonresponse at weeks 1 or 2 predicted subsequent nonresponse at week 4 or later. However, other studies support the idea that early nonresponse predicts subsequent nonresponse and early antipsychotic response predicts future response in first-episode patients, with good specificity and sensitivity.37,38
Overall, treatment response in first-episode schizophrenia is variable. An adequate antipsychotic trial may be longer, 8 to 16 weeks, compared with 4 to 8 weeks in multi-episode patients. Because research suggests that failure to respond to treatment may lead to medication nonadherence,39 it is reasonable to consider switching antipsychotics when a patient experiences minimal or no response to antipsychotic therapy at week 2; however, this should be a patient-specific decision.
How long should you continue therapy after symptom remission?
There is a lack of consensus on the duration of therapy for a patient treated for first-episode schizophrenia because a small percentage (10% to 20%) do not relapse after the first psychotic episode.3 In general, treatment guidelines and expert consensus statements recommend at least 1 to 2 years of treatment before considering a discontinuation trial.7,10-11 Discuss the benefits and risks of maintenance treatment with your patient and obtain informed consent. With patients with minimal insight, obtaining proper consent is not possible and the physician must exercise judgment unilaterally, if necessary, after educating the family.
After at least 12 months of treatment, antipsychotic therapy could continue indefinitely, depending on patient-specific factors. There are no predictors for identifying patients who do not require maintenance therapy beyond the first psychotic episode. The absence of negative and cognitive deficits could provide clues that a patient might be a candidate for antipsychotic tapering.
Predicting the treatment course
Research investigating clinical predictors or biomarkers that forecast whether a patient will respond to treatment is preliminary. Many characteristics have been identified (Table 31,3,4,23,25,40) and include shorter DUP,1 poorer premorbid function,3 antipsychotic discontinuation,3 a trusting patient-doctor relationship,41 and antipsychotic-related adverse effects,23,25 which are predictive of response, nonresponse, relapse, adherence, and nonadherence, respectively.
Bottom Line
The goals of pharmacological treatment of first-episode schizophrenia are to minimize the duration of untreated psychosis and target full remission of positive symptoms using the lowest possible antipsychotic dosages. Pharmacotherapy should continued for 1 to 2 years, with longer duration considered if it is discussed with the patient and with vigilant monitoring for adverse effects and suboptimal medication nonadherence to prevent relapse.
Editor’s note: The second article in this series in the July 2015 issue reviews the rationale and evidence for non-standard, first-line therapies, including long-acting injectable antipsychotics and clozapine.
Related Resources
• Recovery After an Initial Schizophrenia Episode (RAISE) Project Early Treatment Program. National Institute of Mental Health. http://raiseetp.org.
• Martens L, Baker S. Promoting recovery from first episode psychosis: a guide for families. Centre for Addiction and Mental Health. http://www.camh.ca/en/hospital/ Documents/www.camh.net/AboutCAMH/Guideto CAMH/MentalHealthPrograms/SchizophreniaProgram/ 3936PromotingRecoveryFirstEpisodePsychosisfinal.pdf.
Drug Brand Names
Aripiprazole • Abilify Lurasidone • Latuda
Asenapine • Saphris Olanzapine • Zyprexa
Clozapine • Clozaril Paliperidone • Invega
Fluphenazine • Prolixin Quetiapine • Seroquel
Iloperidone • Fanapt Risperidone • Risperdal
Haloperidol • Haldol Ziprasidone • Geodon
Disclosures
Dr. Gardner reports no financial relationships with any companies whose products are mentioned in this article or with manufacturers of competing products.
Dr. Nasrallah is a consultant to Acadia, Alkermes, Lundbeck, Janssen, Merck, Otsuka, and Sunovion, and is a speaker for Alkermes, Lundbeck, Janssen, Otsuka, and Sunovion.
The less time that passes between the onset of psychosis and initiation of appropriate treatment, the greater the patient’s odds of recovery.1 However, relapse prevention is a major clinical challenge because >80% of patients will relapse within 5 years, and, on average, 40% to 50% of patients with a first-episode schizophrenia will relapse within 2 years depending on the definition used and patient characteristics.2 Although there are several explanations and contributing factors to relapses, nonadherence—partial or complete discontinuation of antipsychotics—is a primary risk factor, contributing to a 5-fold increase in relapse risk.3
As such, optimal antipsychotic selection, dosing, and monitoring play an important role in managing this illness. Patients with first-episode psychosis (FEP) are unusual in some ways, compared with patients with multiple episodes of psychosis and represent a different stage of schizophrenia.
In this 2-part series, we will discuss pharmacotherapy for FEP. This article focuses on antipsychotic selection, dosage, and duration of treatment among these patients. The second article, in the July 2015 issue, reviews the rationale and evidence for non-standard, first-line therapies, including long-acting injectable antipsychotics and clozapine.
Defining FEP
FEP refers to a patient who has presented, been evaluated, and received treatment for the first psychotic episode associated with a schizophrenia spectrum diagnosis.4 FEP is part of a trajectory marked by tran sitional periods. The patient transitions from being “healthy” to a prodromal state characterized by: (1) nonpsychotic behavioral disturbances such as depression or obsessive-compulsive disorder, (2) attenuated psychotic symptoms not requiring treatment, then converting to (3) psychotic symptoms prompting initial presentation for antipsychotic pharmacotherapy, leading to (4) a formal diagnosis of schizophreniform disorder and, subsequently, schizophrenia, requiring treatment to stabilize symptoms.
There are 2 critical periods along this continuum: prodromal stage and the duration of untreated psychosis (DUP). The prodromal period is a retrospectively identified time where the patient shows initial nonpsychotic disturbances (eg, cognitive and behavioral symptoms) before exhibiting clinical diagnostic criteria for a schizophrenia spectrum disorder. Approximately one-third of patients exhibiting these symptoms convert to psychosis within 1 year, and early treatment engagement at this stage has been shown to improve outcomes.5 The DUP is the time from when a patient has noticeable psychotic symptoms to initiation of drug treatment. The DUP is a consistent predictor of clinical outcome in schizophrenia, including negative symptoms, quality of life, and functional capacity.1
Antipsychotic selection
Treatment goals for FEP patients include:
• minimizing the DUP
• rapidly stabilizing psychosis
• achieving full symptomatic remission
• preventing relapse.
Several treatment guidelines for managing schizophrenia offer variable recommendations for initial antipsychotic treatment in patients with first-episode schizophrenia (Table 1).6-15 Most recommend second-generation antipsychotics (SGAs) over first-generation antipsychotics (FGAs)6,8,9,13,15 with specific recommendations on minimizing neurologic and metabolic adverse effects—to which FEP patients are susceptible—by avoiding high-potency and neurotoxic FGAs (eg, haloperidol and fluphenazine),7 clozapine,11,14 olanzapine,11 or ziprasidone.14 Two guidelines—the National Institute for Health and Care Excellence and the Scottish Intercollegiate Guidelines Network—do not state a preference for antipsychotic selection.10,12
The rationale for these recommendations is based on efficacy data, tolerability differences, FDA-approved indications, and recent FDA approvals with sparse post-marketing data. Of note, there are a lack of robust data for newer antipsychotics (eg, aripiprazole, paliperidone, iloperidone, asenapine, and lurasidone) in effectively and safely treating FEP; however, given the results of other antipsychotics studies, it is likely the efficacy and tolerability of these drugs can be extrapolated from experience with multi-episode patients.
Study design and demographics. Research studies of FEP share some similarities in study design; however, there is enough variability to make it difficult to compare studies and generalize findings (Table 2).16 The variability of DUP is a limitation when comparing studies because it is a significant predictor of clinical outcome. Patients who abuse substances—and often are more challenging to treat17—typically are excluded from these trials, which could explain the high response rate documented in studies of first-episode schizophrenia.
In addition, some FEP patients included in clinical trials might not be truly antipsychotic naïve; an estimated 25% to 75% of patients in these studies are antipsychotic naïve. This is an important consideration when comparing data on adverse effects that occur early in treatment. Additionally, acknowledging the advantages and disadvantages of how to handle missing data is critical because of the high dropout rate observed in these studies.18
Efficacy. There is a high response rate to antipsychotic therapy—ranging from 46% to 96%, depending on the study—in patients with first-episode schizophrenia.3 The response mainly is seen in reduction of positive symptoms because typically negative and cognitive symptoms do not respond to antipsychotics. One study reported only 29% of patients achieved both positive and negative symptom remission.19 It is likely that secondary negative symptoms caused by social withdrawal, reduced speech, and avoidance improve when positive symptoms subside, but primary negative symptoms endure.In general, there is a lack of evidence suggesting that 1 antipsychotic class or agent is more effective than another. Studies mainly assess effectiveness using the primary outcome measure of all-cause discontinuation, such as the Clinical Antipsychotic Trials of Intervention Effectiveness study.20 This outcome measure is a mixture of patient preference, tolerability, and efficacy that provides a more generalizable gauge on how well the treatment works in the clinic rather than tightly regulated settings such as clinical trials. A recent meta-analysis supports no differences in efficacy among antipsychotics in early-episode psychosis.21
Tolerability. Because there are no significant differences among antipsychotic classes or agents in terms of efficacy in first-episode schizophrenia, drug selection is guided mainly by (1) the adverse effect profile and (2) what should be avoided depending on patient-specific variables. Evidence suggests first-episode patients are more sensitive to adverse effects of antipsychotics, particularly neurologic side effects (see this article at CurrentPsychiatry.com for a table comparing adverse effects of antipsychotics in first-episode psychosis).18,22-29 Overall adverse effect profiles remain similar across FEP or multi-episode patients, but tend to be more exaggerated in drug-naïve patients with FEP.
Regarding FGA side effects, McEvoy et al18 demonstrated the neuroleptic threshold occurs at 50% lower haloperidol dosages in patients with first-episode schizophrenia (2.1 mg/d) compared with multi-episode schizophrenia (4.3 mg/d). Other trials suggest SGAs are associated with a lower risk of extrapyramidal side effects (EPS) or use of adjunctive therapies such as anticholinergic drugs or benzodiazepines.23-27 An exception to this statement is that higher risperidone dosages (≥4 to 6 mg/d) have been found to have higher rates of EPS and use of adjunctive medications to treat these symptoms in FEP.26 This is important because studies report higher discontinuation rates with more severe adverse effects of antipsychotics.
Cardiometabolic effects are of particular concern in first-episode patients because most weight gain happens in the first 3 to 4 months of treatment and remains throughout the first year.18,24,29,30 Studies have shown that olanzapine, quetiapine, and risperidone are associated with more clinically significant weight gain compared with haloperidol and ziprasidone.23-25 Olanzapine-associated weight gain has been reported to be twice that of quetiapine and risperidone.18 Regardless, the EUFEST trial did not find a difference in clinically significant weight gain after 12 months among the antipsychotics studied, including haloperidol and ziprasidone.25
Weight gain associated with these antipsychotics is accompanied by changes in fasting triglycerides, glucose, total cholesterol,23 and high-density lipoprotein cholesterol as well as an increase in body mass index (BMI) categorization29 (eg, shift from normal to overweight).18,25 Patients with lower baseline BMI and in racial minority groups might experience more rapid weight gain regardless of antipsychotic selection.29,30
Hyperprolactinemia could be under-recognized and could contribute to early treatment discontinuation.31 Evidence in patients with first-episode schizophrenia suggests similar outcomes as those seen in multi-episode patients, in whom risperidone is associated with higher prolactin elevations and clinically significant hyperprolactinemia (eg, galactorrhea and gynecomastia) compared with olanzapine, quetiapine, and low-dose haloperidol.18,23,24 However, there is a lack of studies that assess whether long-term therapy with strong D2 receptor antagonists increases the risk of bone demineralization or pathological fractures when started before patients’ bones reach maximum density in their mid-20s.31
Antipsychotic dosing
Given the high rate of treatment response in FEP and patients’ higher sensitivity to antipsychotic adverse effects, particularly EPS, guidelines recommend antipsychotic dosages lower than those used for multi-episode schizophrenia,11 especially FGAs. Based on trial data, commonly used dosages include:
• haloperidol, ≤5 mg/d23-25,29
• olanzapine, 10 mg/d18,23,25,29
• risperidone, ≤4 to 6 mg/d.18,24,29,32
In general, haloperidol and risperidone, 2 to 3 mg/d, were well tolerated and effective in trials. Higher quetiapine dosages of 500 to 600 mg/d could be required.11,18,25
According to a survey on prescribing practices of antipsychotic selection and dosing in first-episode schizophrenia,4 clinical prescribing practices tend to use unnecessarily high initial antipsychotic dosing compared with trial data. There also is variability in the usual target antipsychotic dosage ranging from 50% lower dosages to normal dosages in chronic schizophrenia to above FDA-approved maximum dosages for olanzapine (which may be necessary to counteract tobacco-induced cytochrome P450 1A2 enzyme induction).
In addition, these clinicians reported prescribing aripiprazole, an antipsychotic with weaker evidence (eg, case reports, case series, open-label studies) supporting its efficacy and tolerability in FEP. These prescribing practices could reflect attempts to reduce the DUP and achieve symptom remission, so long as tolerability is not a concern.
Essentially, prescribed dosages should be based on symptom improvement and tolerability. This ideal dosage will vary as illustrated by Kapur et al,33 who reported that FEP patients (N = 20) given haloperidol, 1 mg or 2.5 mg/d, had D2 receptor occupancy rates of 38% to 87%, which was significantly dose-related (1 mg/d mean = 59%, 2.5 mg/d mean = 75%). Clinical response and EPS significantly increased as D2 receptor occupancy exceeded 65% and 78%, respectively.
Antipsychotic response
When should you expect to see symptom improvement in patients with first-episode schizophrenia?
Emsley et al34 reported a 77.6% response rate among first-episode patients (N = 522) treated with low dosages of risperidone (mean modal dosage [MMD] = 3.3 mg/d) and haloperidol (MMD = 2.9 mg/d). They found variable response times that were evenly dispersed over a 10-week period. Nearly one-quarter (22.5%) did not respond until after week 4 and 11.2% did not respond until after week 8. In a study of FEP patients (N = 112) treated with olanzapine (MMD = 11.8 mg/d) or risperidone (MMD = 3.9 mg/d), Gallego et al35 reported a cumulative response of 39.6% at week 8 and 65.1% at week 16.
Although there is evidence that, among multi-episode patients, early nonresponse to antipsychotic therapy could predict subsequent nonresponse,36 the evidence is mixed for first-episode schizophrenia. Studies by Emsley et al34 and Gallego et al35 did not find that early nonresponse at weeks 1 or 2 predicted subsequent nonresponse at week 4 or later. However, other studies support the idea that early nonresponse predicts subsequent nonresponse and early antipsychotic response predicts future response in first-episode patients, with good specificity and sensitivity.37,38
Overall, treatment response in first-episode schizophrenia is variable. An adequate antipsychotic trial may be longer, 8 to 16 weeks, compared with 4 to 8 weeks in multi-episode patients. Because research suggests that failure to respond to treatment may lead to medication nonadherence,39 it is reasonable to consider switching antipsychotics when a patient experiences minimal or no response to antipsychotic therapy at week 2; however, this should be a patient-specific decision.
How long should you continue therapy after symptom remission?
There is a lack of consensus on the duration of therapy for a patient treated for first-episode schizophrenia because a small percentage (10% to 20%) do not relapse after the first psychotic episode.3 In general, treatment guidelines and expert consensus statements recommend at least 1 to 2 years of treatment before considering a discontinuation trial.7,10-11 Discuss the benefits and risks of maintenance treatment with your patient and obtain informed consent. With patients with minimal insight, obtaining proper consent is not possible and the physician must exercise judgment unilaterally, if necessary, after educating the family.
After at least 12 months of treatment, antipsychotic therapy could continue indefinitely, depending on patient-specific factors. There are no predictors for identifying patients who do not require maintenance therapy beyond the first psychotic episode. The absence of negative and cognitive deficits could provide clues that a patient might be a candidate for antipsychotic tapering.
Predicting the treatment course
Research investigating clinical predictors or biomarkers that forecast whether a patient will respond to treatment is preliminary. Many characteristics have been identified (Table 31,3,4,23,25,40) and include shorter DUP,1 poorer premorbid function,3 antipsychotic discontinuation,3 a trusting patient-doctor relationship,41 and antipsychotic-related adverse effects,23,25 which are predictive of response, nonresponse, relapse, adherence, and nonadherence, respectively.
Bottom Line
The goals of pharmacological treatment of first-episode schizophrenia are to minimize the duration of untreated psychosis and target full remission of positive symptoms using the lowest possible antipsychotic dosages. Pharmacotherapy should continued for 1 to 2 years, with longer duration considered if it is discussed with the patient and with vigilant monitoring for adverse effects and suboptimal medication nonadherence to prevent relapse.
Editor’s note: The second article in this series in the July 2015 issue reviews the rationale and evidence for non-standard, first-line therapies, including long-acting injectable antipsychotics and clozapine.
Related Resources
• Recovery After an Initial Schizophrenia Episode (RAISE) Project Early Treatment Program. National Institute of Mental Health. http://raiseetp.org.
• Martens L, Baker S. Promoting recovery from first episode psychosis: a guide for families. Centre for Addiction and Mental Health. http://www.camh.ca/en/hospital/ Documents/www.camh.net/AboutCAMH/Guideto CAMH/MentalHealthPrograms/SchizophreniaProgram/ 3936PromotingRecoveryFirstEpisodePsychosisfinal.pdf.
Drug Brand Names
Aripiprazole • Abilify Lurasidone • Latuda
Asenapine • Saphris Olanzapine • Zyprexa
Clozapine • Clozaril Paliperidone • Invega
Fluphenazine • Prolixin Quetiapine • Seroquel
Iloperidone • Fanapt Risperidone • Risperdal
Haloperidol • Haldol Ziprasidone • Geodon
Disclosures
Dr. Gardner reports no financial relationships with any companies whose products are mentioned in this article or with manufacturers of competing products.
Dr. Nasrallah is a consultant to Acadia, Alkermes, Lundbeck, Janssen, Merck, Otsuka, and Sunovion, and is a speaker for Alkermes, Lundbeck, Janssen, Otsuka, and Sunovion.
1. Perkins DO, Gu H, Boteva K, et al. Relationship between duration of untreated psychosis and outcome in first-episode schizophrenia: a critical review and meta-analysis. Am J Psychiatry. 2005;162(10):1785-1804.
2. Bradford DW, Perkins DO, Lieberman JA. Pharmacological management of first-episode schizophrenia and related nonaffective psychoses. Drugs. 2003;63(21):2265-2283.
3. Robinson D, Woerner MG, Alvir JM, et al. Predictors of relapse following a response from a first episode of schizophrenia or schizoaffective disorder. Arch Gen Psychiatry. 1999;56(3):241-247.
4. Weiden PJ, Buckley PF, Grody M. Understanding and treating “first-episode” schizophrenia. Psychiatr Clin North Am. 2007;30(3):481-510.
5. Madaan V, Bestha DP, Kolli V. Schizophrenia prodrome: an optimal approach. Current Psychiatry. 2014;13(3):16-20, 29-30.
6. Lehman AF, Lieberman JA, Dixon LB, et al; American Psychiatric Association; Steering Committee on Practice Guidelines. Practice guideline for the treatment of patients with schizophrenia, second edition. Am J Psychiatry. 2004;161(suppl 2):1-56.
7. Barnes TR; Schizophrenia Consensus Group of British Association for Psychopharmacology. Evidence-based guidelines for the pharmacological treatment of schizophrenia: recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2011;25(5):567-620.
8. Canadian Psychiatric Association. Clinical practice guideline. Treatment of schizophrenia. Can J Psychiatry. 2005;50(13 suppl 1):7S-57S.
9. McEvoy JP, Scheifler PL, Frances A. Treatment of schizophrenia 1999. Expert consensus guideline series. J Clin Psychiatry. 1999;60(suppl 11):4-80.
10. National Institute for Health and Care Excellence (NICE). Clinical guideline 178: Psychosis and schizophrenia in adults: treatment and management. London, United Kingdom: National Institute for Health and Care Excellence (NICE); 2014.
11. Buchanan RW, Kreyenbuhl J, Kelly DL, et al; Schizophrenia Patient Outcomes Research Team (PORT). The 2009 schizophrenia PORT psychopharmacological treatment recommendations and summary statements. Schizophr Bull. 2010;36(1):71-93.
12. Scottish Intercollegiate Guidelines Network (SIGN). Management of schizophrenia. Edinburgh, Scotland: Scottish Intercollegiate Guidelines Network; 2013. SIGN publication no. 131.
13. Argo TR, Crismon ML, Miller AL, et al. Texas Medication Algorithm Project procedural manual. Schizophrenia treatment algorithms. Austin, Texas: Texas Department of State Health Services; 2008.
14. Marder SR, Essock SM, Miller Al, et al. The Mount Sinai conference on the pharmacotherapy of schizophrenia. Schizophr Bull. 2002;28(1):5-16.
15. Bandelow B, Zohar J, Hollander E, et al; WFSBP Task Force on Treatment Guidelines for Anxiety, Obsessive-Compulsive and Post-Traumatic Stress Disorders. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry. 2008;9(4):248-312.
16. Robinson DG, Woerner MG, Alvir JMJ, et al. Predictors of relapse following response from a first episode of schizophrenia or schizoaffective disorder. Arch Gen Psych. 1999;56(3):241-247.
17. Green AI, Tohen MF, Hamer RM, et al. First episode schizophrenia-related psychosis and substance use disorders: acute response to olanzapine and haloperidol. Schizophr Res. 2004;66(2-3):125-135.
18. McEvoy JP, Lieberman JA, Perkins DO, et al. Efficacy and tolerability of olanzapine, quetiapine, and risperidone in the treatment of early psychosis: a randomized, double-blind 52-week comparison. Am J Psychiatry. 2007;164(7): 1050-1060.
19. Henry LP, Amminger GP, Harris MG, et al. The EPPIC follow-up study of first-episode psychosis: longer-term clinical and functional outcome 7 years after index admission. J Clin Psychiatry. 2010;71(6):716-728.
20. Lieberman JA, Stroup TS, McEvoy JP, et al; Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Investigators. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. New Engl J Med. 2005; 353(12):1209-1223.
21. Crossley NA, Constante M, McGuire P, et al. Efficacy of atypical v. typical antipsychotics in the treatment of early psychosis: meta-analysis. Br J Psychiatry. 2010;196(6):434-439.
22. McEvoy JP, Hogarty GE, Steingard S. Optimal dose of neuroleptic in acute schizophrenia: a controlled study of the neuroleptic threshold and higher haloperidol dose. Arch Gen Psych. 1991;48(8):739-745.
23. Lieberman JA, Tollefson G, Tohen M, et al; HGDH Study Group. Comparative efficacy and safety of atypical and conventional antipsychotic drugs in first-episode psychosis: a randomized, double-blind trial of olanzapine versus haloperidol. Am J Psychiatry. 2003;160(8):1396-1404.
24. Schooler N, Rabinowitz J, Davidson M, et al; Early Psychosis Global Working Group. Risperidone and haloperidol in first-episode psychosis: a long-term randomized trial. Am J Psychiatry. 2005;162(5):947-953.
25. Kahn RS, Fleischhacker WW, Boter H, et al; EUFEST study group. Effectiveness of antipsychotic drugs in first-episode schizophrenia and schizophreniform disorder: an open randomised clinical trial. Lancet. 2008;371(9618):1085-1097.
26. Emsley RA; Risperidone Working Group. Risperidone in the treatment of first-episode psychotic patients: a double-blind multicenter study. Schizophr Bull. 1999;25(4):721-729.
27. Lieberman JA, Phillips M, Gu H, et al. Atypical and conventional antipsychotic drugs in treatment-naïve first-episode schizophrenia: a 52-week randomized trial of clozapine vs chlorpromazine. Neuropsychopharmacology. 2003;28(5):995-1003.
28. Girgis RR, Phillips MR, Li X, et al. Clozapine v. chlorpromazine in treatment-naive, first-episode schizophrenia: 9-year outcomes of a randomised clinical trial. Br J Psychiatry. 2011;199(4):281-288.
29. Robinson DG, Woerner MG, Napolitano B, et al. Randomized comparison of olanzapine versus risperidone for the treatment of first-episode schizophrenia: 4-month outcomes. Am J Psychiatry. 2006;163(12):2096-2102.
30. Zipursky RB, Gu H, Green AI, et al. Course and predictors of weight gain in people with first-episode psychosis treated with olanzapine or haloperidol. Br J Psychiatry. 2005;187:537-543.
31. Taylor M, Waight A, Leonard B. Advances in the understanding and challenges facing the management of first-episode schizophrenia. J Psychopharmacol. 2012; 26(suppl 5):3-5.
32. Merlo MC, Hofer H, Gekle W, et al. Risperidone, 2mg/day vs. 4mg/day, in first-episode, acutely psychotic patients: treatment efficacy and effects on fine motor functioning. J Clin Psychiatry. 2002;63(10):885-891.
33. Kapur S, Zipursky R, Jones C, et al. Relationship between dopamine D2 occupancy, clinical response, and side effects: a double-blind PET study of first-episode schizophrenia. Am J Psychiatry. 2000;157(4):514-520.
34. Emsley R, Rabinowitz J, Medori R. Time course for antipsychotic treatment response in first-episode schizophrenia. Am J Psychiatry. 2006;163(4):743-745.
35. Gallego JA, Robinson DG, Sevy SM, et al. Time to treatment response in first-episode schizophrenia: should acute treatment trials last several months? J Clin Psychiatry. 2011;72(12):1691-1696.
36. Gardner KN, Bostwick JR. Antipsychotic treatment response in schizophrenia. Am J Health Sys Pharm. 2012;69(21):1872-1879.
37. Stauffer VL, Case M, Kinon BJ, et al. Early response to antipsychotic therapy as a clinical marker of subsequent response in the treatment of patients with first-episode psychosis. Psychiatry Res. 2011;187(1-2):42-48.
38. Schennach-Wolff R, Seemüller FH, Mayr A, et al. An early improvement threshold to predict response and remission in first-episode schizophrenia. Br J Psychiatry. 2010;196(6):460-466.
39. Perkins DO, Gu H, Weiden PJ, et al; Comparison of Atypicals in First Episode study group. Predictors of treatment discontinuation and medication nonadherence in patients recovering from a first episode of schizophrenia, schizophreniform disorder, or schizoaffective disorder: a randomized, double-blind, flexible-dose, multicenter study. J Clin Psychiatry. 2008;69(1):106-113.
40. Garner B, Berger GE, Nicolo JP, et al. Pituitary volume and early treatment response in drug-naïve first-episode psychosis patients. Schizophr Res. 2009;113(1):65-71.
41. Sapra M, Weiden PJ, Schooler NR, et al. Reasons for adherence and nonadherence: a pilot study comparing first-and multi-episode schizophrenia patients. Clin Schizophr Relat Psychoses. 2014;7(4):199-206.
1. Perkins DO, Gu H, Boteva K, et al. Relationship between duration of untreated psychosis and outcome in first-episode schizophrenia: a critical review and meta-analysis. Am J Psychiatry. 2005;162(10):1785-1804.
2. Bradford DW, Perkins DO, Lieberman JA. Pharmacological management of first-episode schizophrenia and related nonaffective psychoses. Drugs. 2003;63(21):2265-2283.
3. Robinson D, Woerner MG, Alvir JM, et al. Predictors of relapse following a response from a first episode of schizophrenia or schizoaffective disorder. Arch Gen Psychiatry. 1999;56(3):241-247.
4. Weiden PJ, Buckley PF, Grody M. Understanding and treating “first-episode” schizophrenia. Psychiatr Clin North Am. 2007;30(3):481-510.
5. Madaan V, Bestha DP, Kolli V. Schizophrenia prodrome: an optimal approach. Current Psychiatry. 2014;13(3):16-20, 29-30.
6. Lehman AF, Lieberman JA, Dixon LB, et al; American Psychiatric Association; Steering Committee on Practice Guidelines. Practice guideline for the treatment of patients with schizophrenia, second edition. Am J Psychiatry. 2004;161(suppl 2):1-56.
7. Barnes TR; Schizophrenia Consensus Group of British Association for Psychopharmacology. Evidence-based guidelines for the pharmacological treatment of schizophrenia: recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2011;25(5):567-620.
8. Canadian Psychiatric Association. Clinical practice guideline. Treatment of schizophrenia. Can J Psychiatry. 2005;50(13 suppl 1):7S-57S.
9. McEvoy JP, Scheifler PL, Frances A. Treatment of schizophrenia 1999. Expert consensus guideline series. J Clin Psychiatry. 1999;60(suppl 11):4-80.
10. National Institute for Health and Care Excellence (NICE). Clinical guideline 178: Psychosis and schizophrenia in adults: treatment and management. London, United Kingdom: National Institute for Health and Care Excellence (NICE); 2014.
11. Buchanan RW, Kreyenbuhl J, Kelly DL, et al; Schizophrenia Patient Outcomes Research Team (PORT). The 2009 schizophrenia PORT psychopharmacological treatment recommendations and summary statements. Schizophr Bull. 2010;36(1):71-93.
12. Scottish Intercollegiate Guidelines Network (SIGN). Management of schizophrenia. Edinburgh, Scotland: Scottish Intercollegiate Guidelines Network; 2013. SIGN publication no. 131.
13. Argo TR, Crismon ML, Miller AL, et al. Texas Medication Algorithm Project procedural manual. Schizophrenia treatment algorithms. Austin, Texas: Texas Department of State Health Services; 2008.
14. Marder SR, Essock SM, Miller Al, et al. The Mount Sinai conference on the pharmacotherapy of schizophrenia. Schizophr Bull. 2002;28(1):5-16.
15. Bandelow B, Zohar J, Hollander E, et al; WFSBP Task Force on Treatment Guidelines for Anxiety, Obsessive-Compulsive and Post-Traumatic Stress Disorders. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry. 2008;9(4):248-312.
16. Robinson DG, Woerner MG, Alvir JMJ, et al. Predictors of relapse following response from a first episode of schizophrenia or schizoaffective disorder. Arch Gen Psych. 1999;56(3):241-247.
17. Green AI, Tohen MF, Hamer RM, et al. First episode schizophrenia-related psychosis and substance use disorders: acute response to olanzapine and haloperidol. Schizophr Res. 2004;66(2-3):125-135.
18. McEvoy JP, Lieberman JA, Perkins DO, et al. Efficacy and tolerability of olanzapine, quetiapine, and risperidone in the treatment of early psychosis: a randomized, double-blind 52-week comparison. Am J Psychiatry. 2007;164(7): 1050-1060.
19. Henry LP, Amminger GP, Harris MG, et al. The EPPIC follow-up study of first-episode psychosis: longer-term clinical and functional outcome 7 years after index admission. J Clin Psychiatry. 2010;71(6):716-728.
20. Lieberman JA, Stroup TS, McEvoy JP, et al; Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Investigators. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. New Engl J Med. 2005; 353(12):1209-1223.
21. Crossley NA, Constante M, McGuire P, et al. Efficacy of atypical v. typical antipsychotics in the treatment of early psychosis: meta-analysis. Br J Psychiatry. 2010;196(6):434-439.
22. McEvoy JP, Hogarty GE, Steingard S. Optimal dose of neuroleptic in acute schizophrenia: a controlled study of the neuroleptic threshold and higher haloperidol dose. Arch Gen Psych. 1991;48(8):739-745.
23. Lieberman JA, Tollefson G, Tohen M, et al; HGDH Study Group. Comparative efficacy and safety of atypical and conventional antipsychotic drugs in first-episode psychosis: a randomized, double-blind trial of olanzapine versus haloperidol. Am J Psychiatry. 2003;160(8):1396-1404.
24. Schooler N, Rabinowitz J, Davidson M, et al; Early Psychosis Global Working Group. Risperidone and haloperidol in first-episode psychosis: a long-term randomized trial. Am J Psychiatry. 2005;162(5):947-953.
25. Kahn RS, Fleischhacker WW, Boter H, et al; EUFEST study group. Effectiveness of antipsychotic drugs in first-episode schizophrenia and schizophreniform disorder: an open randomised clinical trial. Lancet. 2008;371(9618):1085-1097.
26. Emsley RA; Risperidone Working Group. Risperidone in the treatment of first-episode psychotic patients: a double-blind multicenter study. Schizophr Bull. 1999;25(4):721-729.
27. Lieberman JA, Phillips M, Gu H, et al. Atypical and conventional antipsychotic drugs in treatment-naïve first-episode schizophrenia: a 52-week randomized trial of clozapine vs chlorpromazine. Neuropsychopharmacology. 2003;28(5):995-1003.
28. Girgis RR, Phillips MR, Li X, et al. Clozapine v. chlorpromazine in treatment-naive, first-episode schizophrenia: 9-year outcomes of a randomised clinical trial. Br J Psychiatry. 2011;199(4):281-288.
29. Robinson DG, Woerner MG, Napolitano B, et al. Randomized comparison of olanzapine versus risperidone for the treatment of first-episode schizophrenia: 4-month outcomes. Am J Psychiatry. 2006;163(12):2096-2102.
30. Zipursky RB, Gu H, Green AI, et al. Course and predictors of weight gain in people with first-episode psychosis treated with olanzapine or haloperidol. Br J Psychiatry. 2005;187:537-543.
31. Taylor M, Waight A, Leonard B. Advances in the understanding and challenges facing the management of first-episode schizophrenia. J Psychopharmacol. 2012; 26(suppl 5):3-5.
32. Merlo MC, Hofer H, Gekle W, et al. Risperidone, 2mg/day vs. 4mg/day, in first-episode, acutely psychotic patients: treatment efficacy and effects on fine motor functioning. J Clin Psychiatry. 2002;63(10):885-891.
33. Kapur S, Zipursky R, Jones C, et al. Relationship between dopamine D2 occupancy, clinical response, and side effects: a double-blind PET study of first-episode schizophrenia. Am J Psychiatry. 2000;157(4):514-520.
34. Emsley R, Rabinowitz J, Medori R. Time course for antipsychotic treatment response in first-episode schizophrenia. Am J Psychiatry. 2006;163(4):743-745.
35. Gallego JA, Robinson DG, Sevy SM, et al. Time to treatment response in first-episode schizophrenia: should acute treatment trials last several months? J Clin Psychiatry. 2011;72(12):1691-1696.
36. Gardner KN, Bostwick JR. Antipsychotic treatment response in schizophrenia. Am J Health Sys Pharm. 2012;69(21):1872-1879.
37. Stauffer VL, Case M, Kinon BJ, et al. Early response to antipsychotic therapy as a clinical marker of subsequent response in the treatment of patients with first-episode psychosis. Psychiatry Res. 2011;187(1-2):42-48.
38. Schennach-Wolff R, Seemüller FH, Mayr A, et al. An early improvement threshold to predict response and remission in first-episode schizophrenia. Br J Psychiatry. 2010;196(6):460-466.
39. Perkins DO, Gu H, Weiden PJ, et al; Comparison of Atypicals in First Episode study group. Predictors of treatment discontinuation and medication nonadherence in patients recovering from a first episode of schizophrenia, schizophreniform disorder, or schizoaffective disorder: a randomized, double-blind, flexible-dose, multicenter study. J Clin Psychiatry. 2008;69(1):106-113.
40. Garner B, Berger GE, Nicolo JP, et al. Pituitary volume and early treatment response in drug-naïve first-episode psychosis patients. Schizophr Res. 2009;113(1):65-71.
41. Sapra M, Weiden PJ, Schooler NR, et al. Reasons for adherence and nonadherence: a pilot study comparing first-and multi-episode schizophrenia patients. Clin Schizophr Relat Psychoses. 2014;7(4):199-206.
When it’s time for ‘the talk’: Sexuality and your geriatric patient
Recent studies suggest that most older adults maintain sexual interest well into late life; many, however, experience sexual dysfunction. This article provides psychiatric practitioners with current information regarding sexuality and aging, as well as psychiatric and systemic medical comorbidities and sexual side effects of medications. Practice guidelines for assessing and managing sexual dysfunction have been developed for use in many medical specialties, and such guidance would be welcome in psychiatric practice.
This article addresses the myth of “geriatric asexuality” and its potential impact on clinical practice, the effects of age-related physiological changes on sexual activity, the importance of sexuality in the lives of older adults, and sensitive questions clinicians can pose about geriatric sexuality. We also will discuss:
• the importance of including a sexual assessment in the comprehensive psychiatric evaluation
• recognizing sexual dysfunction
• providing appropriate management within a multi-disciplinary, collaborative approach.
Sexuality after 65
Regardless of age, sexual activity can provide a sense of comfort and elicit a positive emotional and physical response.1 Hillman2 defined human sexuality as any combination of sexual behavior, emotional intimacy, and sense of sexual identity.
Sexuality in the aging population generally is an understudied area, obscured by the myth of “geriatric asexuality” and subject to numerous psychosocial variables.1 Previous research, focused on a biological perspective of sexuality, has largely overlooked psychological and social influences.3 It has been assumed that, with age, physical and hormonal changes or chronic illness ordinarily reduce or eliminate sexual desire and sexual behavior.3 However, the majority of older adults (defined as age ≥65) report a moderate-to-high level of sexual interest well into late life.1,3
Sexual function remains a subject often neglected in psychiatry. Sexual dysfunctions, as described in the DSM-5,4 do not include age-related changes in sexual function. In addition to physiological changes, sexual difficulties can result from relationship strain, systemic medical or psychiatric disorders, and sexual side effects of medications.
CASE REPORT
Mr. C, age 71 and married, is being treated for a major depressive episode that followed a course of shingles and persistent postherpetic neuralgia. Medications are: escitalopram, 20 mg/d; pregabalin, 150 mg/d; and ramipril, 5 mg/d. Mr. C is physically active and involved in social activities; he has no substance use history. He attends clinic visits with his wife.
Mr. C reports that despite significant improvement of his depressive and pain symptoms, he now experiences sexual difficulties, which he seems hesitant to discuss in detail. According to his wife, Mr. C appears to lack sexual desire and has difficulty initiating and maintaining an erection. She asks Mr. C’s psychiatrist whether she should stop her estrogen treatment, intended to enhance her sexual function, given that the couple is no longer engaging in sexual intercourse.
Mr. C admits to missing physical intimacy; however, he states, “If I have to make a choice between having sex with my wife and getting this depression out of my head, I’m going to pick getting rid of the depression.” Mrs. C says she is becoming dissatisfied with their marriage and the limited time she and her husband now spend together. Mr. C’s psychiatrist suggests that Mr. C and his wife undergo couples counseling.
Physiological changes with aging
In both women and men, the reproductive system undergoes age-related physiological changes.
Women. In women, the phase of decline in ovarian function and resulting decline in sex steroid production (estradiol and progesterone) is referred to as the climacteric, with menopause being determined retrospectively by the cessation of a menstrual period for 1 year.5
Menopausal symptoms typically occur between age 40 and 58; the average age of menopause is 51.6,7 Both estradiol and progesterone levels decline with menopause, and anovulation and ovarian failure ensue. A more gradual decline of female testosterone levels also occurs with aging, starting in the fourth decade of life.8
Clinical manifestations of menopause include vasomotor symptoms (ie, “hot flushes”), sleep disturbances, anxiety and depressive symptoms, decreased bone mineral density, and increased risk of cardiovascular disease.6,7 Loss of estrogen as well as continued loss of testosterone can result in dyspareunia because of atrophy and decreased vulvar and vaginal lubrication, with sexual excitement achieved less quickly, and a decreased intensity of orgasm.7
Men. Research has shown that testosterone levels are highest in men in the second and third decades, with a subsequent gradual decline.9 Older men with a low testosterone level are described as experiencing “late-onset hypogonadism,” also known by the popularized term “andropause.”10 This is attributed to decreased activity at the testicular and hypothalamic levels.10
Nonetheless, only a small fraction of older men with confirmed androgen deficiency are clinically symptomatic.11,12 Low testosterone is associated with decreased libido; it can hinder morning erections, contribute to erectile dysfunction, and result in erections that require physical stimulation.13
Notably, erectile dysfunction involves several other etiologic factors: psychiatric (eg, relationship difficulties, depression), neurogenic (eg, spinal cord injury), endocrine (eg, hyperprolactinemia), arteriogenic (eg, hypertension, type 2 diabetes mellitus), and drug-induced (eg, antidepressants, antihypertensives).14 A low testosterone level also has been associated with potential cognitive changes, decreased bone mineral density, metabolic syndrome (eg, increased risk of type 2 diabetes mellitus), and cardiovascular mortality.10
Effects on sexual activity. How much age-related physiological changes impact sexual practices in the geriatric population is uncertain. A study following 3,302 women through menopause over 6 years found some decline in sexual activity; however, this decline was not associated with increased sexual pain, decreased desire, or lack of arousal.15 Men continue to find ways to remain sexually active despite physiological changes (eg, erectile difficulties), but the etiology of sexual dysfunction in later life remains multi-modal, involving physical, psychological, and relational factors.16,17
Sexual practices in older adults
Researchers for the National Social Life, Health, and Aging Project (NSHAP) have examined sexual activities, behaviors, and problems in >3,000 older community-dwelling men and women across the United States, using information collected from in-home interviews.18 This study found that sexual activity, defined as any mutually voluntary sexual contact with another person, declines with age; however, even in the oldest age group (age 75 to 85), 39% of men and 17% of women reported being sexually active in the last 12 months. Among these persons, 54% reported sexual activity at least 2 times per month; 23% reported having sex at least once a week; and 32% reported engaging in oral sex. Partner availability predicted sexual activity.
Respondents with self-reported poor physical health were more likely to experience sexual problems (eg, difficulty with erection or lubrication, dyspareunia, and lack of pleasure). The most commonly reported reason for sexual inactivity in those with a spouse or other intimate partner was the male partner’s poor physical health.18
A longitudinal study, part of the Women’s Healthy Ageing Project, examined changes in sexual function at late menopause compared with early menopause. Although the researchers also found an age-related decrease in sexual activity, 50% of their late-menopause respondents (mean age, 70; range, 64 to 77) still reported sexual activity in the previous month, with 35% of this subgroup reporting sexual activity at least once a week; 83% reported sexual thoughts or fantasies.19 Availability of a partner, absence of a history of depression, moderate (compared with no) alcohol consumption, and better cognitive function were significantly associated with a higher level of sexual activity.19
In the Successful Aging Evaluation study, conducted in San Diego County, California, community-dwelling older partnered adults age 50 to 99 (mean age, 75) were surveyed about their sexual health after a cognitive screen by telephone20; rating scales for depression, anxiety, and physical function also were included. Results included 41% of men and 35% of women reporting sexual activity at least once a week, and only 21% of men and 24% of women reporting no sexual activity in the previous year. Depressive symptoms were most highly correlated with lack of sexual activity.20
Overall, these studies reveal that positive physical and mental health, access to a healthy partner, and a positive attitude toward sex are correlated with sexual activity in later life, whereas barriers to sexual activity include lack of a healthy sexual partner, depression, and chronic systemic medical illnesses, such as coronary artery disease or type 2 diabetes mellitus.13,17,21-24 Sexual activity and satisfaction have been positively associated with perceived general well-being and self-esteem.25,26 Conversely, sexual difficulties secondary to disease can be a source of distress for couples.27
Possibly overlooked? It is important to note that sexuality itself is a subjective area. Psychological intimacy is a universal phenomenon, and its physical expression may evolve as couples accommodate to age-related bodily changes. Means of achieving physical closeness, other than intercourse (eg, intimate touching, hand holding, kissing, or even acts of caretaking), may not be adequately captured in studies that look specifically at sexual activity.
Taking a sexual history in a geriatric patient
Because sexuality can be an uncomfortable topic for geriatric patients to discuss, sexual problems in this population often go unrecognized. It has been suggested that psychiatrists are more likely to inquire about sexual activity in middle-aged patients than geriatric patients with the same psychiatric presentation—perhaps illustrating a bias against taking a sexual history from a geriatric patient.28 However, because many older patients can experience depression or anxiety disorders in relation to normal sexual changes or sexual dysfunction within the context of their intimate relationships, it is essential to bring these issues to light.
Although a sexual history may not be the focus of a first clinical encounter, consider making such an assessment at a relatively early stage of patient care. The importance of such dialogue is 2-fold:
• It demonstrates to the patient that talking about sexuality in a respectful and empathic manner is appropriate and can encourage patients to communicate more effectively about sexuality with clinicians and with sexual partners.
• It helps elicit medical information needed to make an accurate diagnosis and provide adequate management.
How to begin. As a starting point to taking a sexual history, an open-ended invitation for the geriatric patient to share information may be best, such as “What would you like to tell me about your sexual life?” See further suggestions (Table 1) and examples of more detailed questions to ask once a dialogue has been initiated (Table 2). Additional factors that may contribute to sexual dysfunction are presented in Table 3.1,27,29,30
CASE CONTINUED
In Mr. C’s case, an assessment of his sexual history, including risk factors for sexual dysfunction, is completed. Results from laboratory investigations, including a total testosterone level, are within normal limits.
Mr. C asks about using medications with fewer sexual side effects (he has been taking 3 medications that can contribute to sexual dysfunction). A gradual cross-taper of escitalopram, 20 mg/d, to mirtazapine, 45 mg/d, is implemented, along with tapering pregabalin to 50 mg/d.
Mr. C’s psychiatric and pain symptom improvement is maintained. He notices a boost in his sexual desire but has minimal improvement in erectile dysfunction. He is encouraged to speak with his primary care physician about an antihypertensive agent with less impact on sexual function, as well as therapeutic agents for erectile dysfunction; these, he declines.
At a subsequent visit, Mr. C reports feeling less apprehension about sexual performance. He is now willing to consider further medication options with his primary care physician, and agrees to a recommendation for couples psychotherapy.
As illustrated in Mr. C’s case, the recommended sexual assessment and management strategies to consider at a minimum in psychiatric practice are listed in Table 4.
STI risk in geriatric patients
The risk of sexually transmitted infections (STIs), including human immunodeficiency virus (HIV), often is overlooked in sexually active older adults. Although STIs are more common among younger adults, there is recent evidence of increased incidence in the geriatric population31 (with the highest risk of incident HIV and some STIs in older men who have sex with men32). These increased rates can be explained, at least in part, by:
• older men being less likely to use a condom during sexual activity
• promotion of viral entry in older women through a drier, thinner vaginal wall
• increased longevity of HIV-positive persons.31
Routine STI screening is not warranted in all older adults, but education and prevention strategies in sexually active seniors who are at greater risk of STIs are recommended. Particularly, clinicians should seek opportunities to discuss risk factors and safe sex practices (eg, using condoms, limiting number of sexual partners, practicing good hygiene, engaging in preventive care), and provide behavioral counseling where appropriate.31,33
Additional considerations in geriatric sexuality
Because psychiatric and systemic medical conditions can hinder sexual function, it is essential to identify and manage these conditions. Several neuropsychiatric disorders, including mood and neurocognitive disorders, can not only cause sexual dysfunction, but also can raise ethical dilemmas for clinicians, such as reduced decisional capacity in cognitively impaired patients to consent to sexual activity.1,34
In some patients, psychological, environmental, and pharmacological treatment options may help. A phosphodiesterase type 5 inhibitor for erectile dysfunction can be prescribed by the primary care physician, a psychiatrist, or another specialist, depending on the physician’s expertise and comfort level.
Sequencing of sexual dysfunction. Notably, there is a common paradox in mood disorders. Decreased sexual interest or performance may represent an aspect of anhedonia associated with depression, whereas sexual dysfunction could also result from medication use (particularly that of serotonergic antidepressants, such as selective serotonin reuptake inhibitors and serotonin-norepinephrine inhibitors), even as other depressive symptoms improve. Therefore, it is critical to analyze sequencing of sexual dysfunction—as part of the presenting mood symptoms or dysfunction induced by antidepressant treatment.
Geriatric sexuality in the digital age. Because older adults represent a rapidly growing segment of digital device users,35 Internet use is likely to play a role in the future of sexuality and “digital intimacy,” in that older adults can engage in online sexual activities. The Internet also can be a tool to access medical education.
Related Resources
• Burghardt KJ, Gardner KN. Sildenafil for SSRI-induced sexual dysfunction. Current Psychiatry. 2013;12(4):29-32,A.
• Maciel M, Laganà L. Older women’s sexual desire problems: biopsychosocial factors impacting them and barriers to their clinical assessment [published online January 5, 2014]. Biomed Res Int. 2014;2014:107217. doi: 10.1155/2014/107217.
Drug Brand Names
Bupropion • Wellbutrin, Zyban Mirtazapine • Remeron
Carbamazepine • Tegretol Oxcarbazepine • Trileptal
Clonidine • Catapres Phenobarbital • Luminal
Donepezil • Aricept Phenytoin • Dilantin
Escitalopram • Lexapro Pregabalin • Lyrica
Gabapentin • Neurontin Ramipril • Altace
Lamotrigine • Lamictal Rivastigmine • Exelon
Lithium • Eskalith, Lithobid Trazodone • Desyrel
Memantine • Namenda Valproic acid • Depakote
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Jagus CE, Benbow SM. Sexuality in older men with mental health problems. Sex Relation Ther. 2002;17(3):271-279.
2. Hillman JL. Clinical perspectives on elderly sexuality. New York, NY: Springer; 2000.
3. DeLamater JD, Sill M. Sexual desire in later life. J Sex Res. 2005;42(2):138-149.
4. Diagnostic and statistical manual of mental disorders. 5th ed. Washington, DC: American Psychiatric Association; 2013.
5. Laufer LR, Gambone JC. Climacteric: menopause and peri-and postmenopause. In: Hacker NF, Gambone JC, Hobel CJ. Hacker and Moore’s essentials of obstetrics and gynecology. 5th ed. Philadelphia, PA: Saunders/Elsevier; 2010:379-385.
6. Wilson MM. Menopause. Clin Geriatr Med. 2003;19(3): 483-506.
7. Reid R, Abramson BL, Blake J, et al. Managing menopause. J Obstet Gynaecol Can. 2014;36(9):830-838.
8. Horstman AM, Dillon EL, Urban RJ, et al. The role of androgens and estrogens on healthy aging and longevity. J Gerontol A Biol Sci Med Sci. 2012;67(11):1140-1152.
9. Wu FC, Tajar A, Pye SR, et al. Hypothalamic-pituitary-testicular axis disruptions in older men are differentially linked to age and modifiable risk factors: the European Male Aging Study. J Clin Endocrinol Metab. 2008;93(7):2737-2745.
10. Basaria S. Reproductive aging in men. Endocrinol Metab Clin North Am. 2013;42(2):255-270.
11. Wu FC, Tajar A, Beynon JM, et al. Identification of late-onset hypogonadism in middle-aged and elderly men. N Engl J Med. 2010;363(2):123-135.
12. Araujo AB, Esche GR, Kupelian V, et al. Prevalence of symptomatic androgen deficiency in men. J Clin Endocrinol Metab. 2007;92(11):4241-4247.
13. Lochlainn MN, Kenny RA. Sexual activity and aging. J Am Med Dir Assoc. 2013;14(8):565-572.
14. McMahon CG. Erectile dysfunction. Intern Med J. 2014;44(1):18-26.
15. Avis NE, Brockwell S, Randolph JF Jr, et al. Longitudinal changes in sexual functioning as women transition through menopause: results from the Study of Women’s Health Across the Nation. Menopause. 2009;16(3):442-452.
16. Perelman M, Shabsigh R, Seftel A, et al. Attitudes of men with erectile dysfunction: a cross-national survey. J Sex Med. 2005;2(3):397-406.
17. Corona G, Rastrelli G, Maseroli E, et al. Sexual function of the ageing male. Best Pract Res Clin Endocrinol Metab. 2013;27(4):581-601.
18. Lindau ST, Schumm LP, Laumann EO, et al. A study of sexuality and health among older adults in the United States. N Engl J Med. 2007;357(8):762-774.
19. Lonnèe-Hoffmann RA, Dennerstein L, Lehert P, et al. Sexual function in the late postmenopause: a decade of follow-up in a population-based cohort of Australian women. J Sex Med. 2014;11(8):2029-2038.
20. Wang V, Depp CA, Ceglowski J, et al. Sexual health and function in later life: a population-based study of 606 older adults with a partner. Am J Geriatr Psychiatry. 2015;23(3):227-233.
21. Garrett D. Psychosocial barriers to sexual intimacy for older people. Br J Nurs. 2014;23(6):327-331.
22. DeLamater J, Karraker A. Sexual functioning in older adults. Curr Psychiatry Rep. 2009;11(1):6-11.
23. DeLamater J. Sexual expression in later life: a review and synthesis. J Sex Res. 2012;49(2-3):125-141.
24. Inelmen EM, Sergi G, Girardi A, et al. The importance of sexual health in the elderly: breaking down barriers and taboos. Aging Clin Exp Res. 2012;24(suppl 3):31-34.
25. Choi KB, Jang SH, Lee MY, et al. Sexual life and self-esteem in married elderly. Arch Gerontol Geriatr. 2011;53(1):e17-e20.
26. Davison SL, Bell RJ, LaChina M, et al. The relationship between self-reported sexual satisfaction and general well-being in women. J Sex Med. 2009;6(10):2690-2697.
27. Morley JE, Tariq SH. Sexuality and disease. Clin Geriatr Med. 2003;19(3):563-573.
28. Bouman WP, Arcelus J. Are psychiatrists guilty of “ageism” when it comes to taking a sexual history? Int J Geriatr Psychiatry. 2001;16(1):27-31.
29. La Torre A, Giupponi G, Duffy DM, et al. Sexual dysfunction related to psychotropic drugs: a critical review. Part III: mood stabilizers and anxiolytic drugs. Pharmacopsychiatry. 2014;47(1):1-6.
30. Tucker I. Management of inappropriate sexual behaviors in dementia: a literature review. Int Psychogeriatr. 2010; 22(5):683-692.
31. Imparato T, Sanders D. STD prevalence demands clinical awareness. Aging Well. 2012;5(1):14.
32. Poynten IM, Grulich AE, Templeton DJ. Sexually transmitted infections in older populations. Curr Opin Infect Dis. 2013;26(1):80-85.
33. Talashek ML, Tichy AM, Epping H. Sexually transmitted diseases in the elderly—issues and recommendations. J Gerontol Nurs. 1990;16(4):33-40.
34. Benbow SM, Jagus CE. Sexuality in older women with mental health problems. Sex Relation Ther. 2002;17(3):261-270.
35. Veenhof B, Timusk P. Online activities of Canadian boomers and seniors. http://www.statcan.gc.ca/pub/ 11-008-x/2009002/article/10910-eng.htm#tphp. Accessed March 26, 2015.
Recent studies suggest that most older adults maintain sexual interest well into late life; many, however, experience sexual dysfunction. This article provides psychiatric practitioners with current information regarding sexuality and aging, as well as psychiatric and systemic medical comorbidities and sexual side effects of medications. Practice guidelines for assessing and managing sexual dysfunction have been developed for use in many medical specialties, and such guidance would be welcome in psychiatric practice.
This article addresses the myth of “geriatric asexuality” and its potential impact on clinical practice, the effects of age-related physiological changes on sexual activity, the importance of sexuality in the lives of older adults, and sensitive questions clinicians can pose about geriatric sexuality. We also will discuss:
• the importance of including a sexual assessment in the comprehensive psychiatric evaluation
• recognizing sexual dysfunction
• providing appropriate management within a multi-disciplinary, collaborative approach.
Sexuality after 65
Regardless of age, sexual activity can provide a sense of comfort and elicit a positive emotional and physical response.1 Hillman2 defined human sexuality as any combination of sexual behavior, emotional intimacy, and sense of sexual identity.
Sexuality in the aging population generally is an understudied area, obscured by the myth of “geriatric asexuality” and subject to numerous psychosocial variables.1 Previous research, focused on a biological perspective of sexuality, has largely overlooked psychological and social influences.3 It has been assumed that, with age, physical and hormonal changes or chronic illness ordinarily reduce or eliminate sexual desire and sexual behavior.3 However, the majority of older adults (defined as age ≥65) report a moderate-to-high level of sexual interest well into late life.1,3
Sexual function remains a subject often neglected in psychiatry. Sexual dysfunctions, as described in the DSM-5,4 do not include age-related changes in sexual function. In addition to physiological changes, sexual difficulties can result from relationship strain, systemic medical or psychiatric disorders, and sexual side effects of medications.
CASE REPORT
Mr. C, age 71 and married, is being treated for a major depressive episode that followed a course of shingles and persistent postherpetic neuralgia. Medications are: escitalopram, 20 mg/d; pregabalin, 150 mg/d; and ramipril, 5 mg/d. Mr. C is physically active and involved in social activities; he has no substance use history. He attends clinic visits with his wife.
Mr. C reports that despite significant improvement of his depressive and pain symptoms, he now experiences sexual difficulties, which he seems hesitant to discuss in detail. According to his wife, Mr. C appears to lack sexual desire and has difficulty initiating and maintaining an erection. She asks Mr. C’s psychiatrist whether she should stop her estrogen treatment, intended to enhance her sexual function, given that the couple is no longer engaging in sexual intercourse.
Mr. C admits to missing physical intimacy; however, he states, “If I have to make a choice between having sex with my wife and getting this depression out of my head, I’m going to pick getting rid of the depression.” Mrs. C says she is becoming dissatisfied with their marriage and the limited time she and her husband now spend together. Mr. C’s psychiatrist suggests that Mr. C and his wife undergo couples counseling.
Physiological changes with aging
In both women and men, the reproductive system undergoes age-related physiological changes.
Women. In women, the phase of decline in ovarian function and resulting decline in sex steroid production (estradiol and progesterone) is referred to as the climacteric, with menopause being determined retrospectively by the cessation of a menstrual period for 1 year.5
Menopausal symptoms typically occur between age 40 and 58; the average age of menopause is 51.6,7 Both estradiol and progesterone levels decline with menopause, and anovulation and ovarian failure ensue. A more gradual decline of female testosterone levels also occurs with aging, starting in the fourth decade of life.8
Clinical manifestations of menopause include vasomotor symptoms (ie, “hot flushes”), sleep disturbances, anxiety and depressive symptoms, decreased bone mineral density, and increased risk of cardiovascular disease.6,7 Loss of estrogen as well as continued loss of testosterone can result in dyspareunia because of atrophy and decreased vulvar and vaginal lubrication, with sexual excitement achieved less quickly, and a decreased intensity of orgasm.7
Men. Research has shown that testosterone levels are highest in men in the second and third decades, with a subsequent gradual decline.9 Older men with a low testosterone level are described as experiencing “late-onset hypogonadism,” also known by the popularized term “andropause.”10 This is attributed to decreased activity at the testicular and hypothalamic levels.10
Nonetheless, only a small fraction of older men with confirmed androgen deficiency are clinically symptomatic.11,12 Low testosterone is associated with decreased libido; it can hinder morning erections, contribute to erectile dysfunction, and result in erections that require physical stimulation.13
Notably, erectile dysfunction involves several other etiologic factors: psychiatric (eg, relationship difficulties, depression), neurogenic (eg, spinal cord injury), endocrine (eg, hyperprolactinemia), arteriogenic (eg, hypertension, type 2 diabetes mellitus), and drug-induced (eg, antidepressants, antihypertensives).14 A low testosterone level also has been associated with potential cognitive changes, decreased bone mineral density, metabolic syndrome (eg, increased risk of type 2 diabetes mellitus), and cardiovascular mortality.10
Effects on sexual activity. How much age-related physiological changes impact sexual practices in the geriatric population is uncertain. A study following 3,302 women through menopause over 6 years found some decline in sexual activity; however, this decline was not associated with increased sexual pain, decreased desire, or lack of arousal.15 Men continue to find ways to remain sexually active despite physiological changes (eg, erectile difficulties), but the etiology of sexual dysfunction in later life remains multi-modal, involving physical, psychological, and relational factors.16,17
Sexual practices in older adults
Researchers for the National Social Life, Health, and Aging Project (NSHAP) have examined sexual activities, behaviors, and problems in >3,000 older community-dwelling men and women across the United States, using information collected from in-home interviews.18 This study found that sexual activity, defined as any mutually voluntary sexual contact with another person, declines with age; however, even in the oldest age group (age 75 to 85), 39% of men and 17% of women reported being sexually active in the last 12 months. Among these persons, 54% reported sexual activity at least 2 times per month; 23% reported having sex at least once a week; and 32% reported engaging in oral sex. Partner availability predicted sexual activity.
Respondents with self-reported poor physical health were more likely to experience sexual problems (eg, difficulty with erection or lubrication, dyspareunia, and lack of pleasure). The most commonly reported reason for sexual inactivity in those with a spouse or other intimate partner was the male partner’s poor physical health.18
A longitudinal study, part of the Women’s Healthy Ageing Project, examined changes in sexual function at late menopause compared with early menopause. Although the researchers also found an age-related decrease in sexual activity, 50% of their late-menopause respondents (mean age, 70; range, 64 to 77) still reported sexual activity in the previous month, with 35% of this subgroup reporting sexual activity at least once a week; 83% reported sexual thoughts or fantasies.19 Availability of a partner, absence of a history of depression, moderate (compared with no) alcohol consumption, and better cognitive function were significantly associated with a higher level of sexual activity.19
In the Successful Aging Evaluation study, conducted in San Diego County, California, community-dwelling older partnered adults age 50 to 99 (mean age, 75) were surveyed about their sexual health after a cognitive screen by telephone20; rating scales for depression, anxiety, and physical function also were included. Results included 41% of men and 35% of women reporting sexual activity at least once a week, and only 21% of men and 24% of women reporting no sexual activity in the previous year. Depressive symptoms were most highly correlated with lack of sexual activity.20
Overall, these studies reveal that positive physical and mental health, access to a healthy partner, and a positive attitude toward sex are correlated with sexual activity in later life, whereas barriers to sexual activity include lack of a healthy sexual partner, depression, and chronic systemic medical illnesses, such as coronary artery disease or type 2 diabetes mellitus.13,17,21-24 Sexual activity and satisfaction have been positively associated with perceived general well-being and self-esteem.25,26 Conversely, sexual difficulties secondary to disease can be a source of distress for couples.27
Possibly overlooked? It is important to note that sexuality itself is a subjective area. Psychological intimacy is a universal phenomenon, and its physical expression may evolve as couples accommodate to age-related bodily changes. Means of achieving physical closeness, other than intercourse (eg, intimate touching, hand holding, kissing, or even acts of caretaking), may not be adequately captured in studies that look specifically at sexual activity.
Taking a sexual history in a geriatric patient
Because sexuality can be an uncomfortable topic for geriatric patients to discuss, sexual problems in this population often go unrecognized. It has been suggested that psychiatrists are more likely to inquire about sexual activity in middle-aged patients than geriatric patients with the same psychiatric presentation—perhaps illustrating a bias against taking a sexual history from a geriatric patient.28 However, because many older patients can experience depression or anxiety disorders in relation to normal sexual changes or sexual dysfunction within the context of their intimate relationships, it is essential to bring these issues to light.
Although a sexual history may not be the focus of a first clinical encounter, consider making such an assessment at a relatively early stage of patient care. The importance of such dialogue is 2-fold:
• It demonstrates to the patient that talking about sexuality in a respectful and empathic manner is appropriate and can encourage patients to communicate more effectively about sexuality with clinicians and with sexual partners.
• It helps elicit medical information needed to make an accurate diagnosis and provide adequate management.
How to begin. As a starting point to taking a sexual history, an open-ended invitation for the geriatric patient to share information may be best, such as “What would you like to tell me about your sexual life?” See further suggestions (Table 1) and examples of more detailed questions to ask once a dialogue has been initiated (Table 2). Additional factors that may contribute to sexual dysfunction are presented in Table 3.1,27,29,30
CASE CONTINUED
In Mr. C’s case, an assessment of his sexual history, including risk factors for sexual dysfunction, is completed. Results from laboratory investigations, including a total testosterone level, are within normal limits.
Mr. C asks about using medications with fewer sexual side effects (he has been taking 3 medications that can contribute to sexual dysfunction). A gradual cross-taper of escitalopram, 20 mg/d, to mirtazapine, 45 mg/d, is implemented, along with tapering pregabalin to 50 mg/d.
Mr. C’s psychiatric and pain symptom improvement is maintained. He notices a boost in his sexual desire but has minimal improvement in erectile dysfunction. He is encouraged to speak with his primary care physician about an antihypertensive agent with less impact on sexual function, as well as therapeutic agents for erectile dysfunction; these, he declines.
At a subsequent visit, Mr. C reports feeling less apprehension about sexual performance. He is now willing to consider further medication options with his primary care physician, and agrees to a recommendation for couples psychotherapy.
As illustrated in Mr. C’s case, the recommended sexual assessment and management strategies to consider at a minimum in psychiatric practice are listed in Table 4.
STI risk in geriatric patients
The risk of sexually transmitted infections (STIs), including human immunodeficiency virus (HIV), often is overlooked in sexually active older adults. Although STIs are more common among younger adults, there is recent evidence of increased incidence in the geriatric population31 (with the highest risk of incident HIV and some STIs in older men who have sex with men32). These increased rates can be explained, at least in part, by:
• older men being less likely to use a condom during sexual activity
• promotion of viral entry in older women through a drier, thinner vaginal wall
• increased longevity of HIV-positive persons.31
Routine STI screening is not warranted in all older adults, but education and prevention strategies in sexually active seniors who are at greater risk of STIs are recommended. Particularly, clinicians should seek opportunities to discuss risk factors and safe sex practices (eg, using condoms, limiting number of sexual partners, practicing good hygiene, engaging in preventive care), and provide behavioral counseling where appropriate.31,33
Additional considerations in geriatric sexuality
Because psychiatric and systemic medical conditions can hinder sexual function, it is essential to identify and manage these conditions. Several neuropsychiatric disorders, including mood and neurocognitive disorders, can not only cause sexual dysfunction, but also can raise ethical dilemmas for clinicians, such as reduced decisional capacity in cognitively impaired patients to consent to sexual activity.1,34
In some patients, psychological, environmental, and pharmacological treatment options may help. A phosphodiesterase type 5 inhibitor for erectile dysfunction can be prescribed by the primary care physician, a psychiatrist, or another specialist, depending on the physician’s expertise and comfort level.
Sequencing of sexual dysfunction. Notably, there is a common paradox in mood disorders. Decreased sexual interest or performance may represent an aspect of anhedonia associated with depression, whereas sexual dysfunction could also result from medication use (particularly that of serotonergic antidepressants, such as selective serotonin reuptake inhibitors and serotonin-norepinephrine inhibitors), even as other depressive symptoms improve. Therefore, it is critical to analyze sequencing of sexual dysfunction—as part of the presenting mood symptoms or dysfunction induced by antidepressant treatment.
Geriatric sexuality in the digital age. Because older adults represent a rapidly growing segment of digital device users,35 Internet use is likely to play a role in the future of sexuality and “digital intimacy,” in that older adults can engage in online sexual activities. The Internet also can be a tool to access medical education.
Related Resources
• Burghardt KJ, Gardner KN. Sildenafil for SSRI-induced sexual dysfunction. Current Psychiatry. 2013;12(4):29-32,A.
• Maciel M, Laganà L. Older women’s sexual desire problems: biopsychosocial factors impacting them and barriers to their clinical assessment [published online January 5, 2014]. Biomed Res Int. 2014;2014:107217. doi: 10.1155/2014/107217.
Drug Brand Names
Bupropion • Wellbutrin, Zyban Mirtazapine • Remeron
Carbamazepine • Tegretol Oxcarbazepine • Trileptal
Clonidine • Catapres Phenobarbital • Luminal
Donepezil • Aricept Phenytoin • Dilantin
Escitalopram • Lexapro Pregabalin • Lyrica
Gabapentin • Neurontin Ramipril • Altace
Lamotrigine • Lamictal Rivastigmine • Exelon
Lithium • Eskalith, Lithobid Trazodone • Desyrel
Memantine • Namenda Valproic acid • Depakote
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
Recent studies suggest that most older adults maintain sexual interest well into late life; many, however, experience sexual dysfunction. This article provides psychiatric practitioners with current information regarding sexuality and aging, as well as psychiatric and systemic medical comorbidities and sexual side effects of medications. Practice guidelines for assessing and managing sexual dysfunction have been developed for use in many medical specialties, and such guidance would be welcome in psychiatric practice.
This article addresses the myth of “geriatric asexuality” and its potential impact on clinical practice, the effects of age-related physiological changes on sexual activity, the importance of sexuality in the lives of older adults, and sensitive questions clinicians can pose about geriatric sexuality. We also will discuss:
• the importance of including a sexual assessment in the comprehensive psychiatric evaluation
• recognizing sexual dysfunction
• providing appropriate management within a multi-disciplinary, collaborative approach.
Sexuality after 65
Regardless of age, sexual activity can provide a sense of comfort and elicit a positive emotional and physical response.1 Hillman2 defined human sexuality as any combination of sexual behavior, emotional intimacy, and sense of sexual identity.
Sexuality in the aging population generally is an understudied area, obscured by the myth of “geriatric asexuality” and subject to numerous psychosocial variables.1 Previous research, focused on a biological perspective of sexuality, has largely overlooked psychological and social influences.3 It has been assumed that, with age, physical and hormonal changes or chronic illness ordinarily reduce or eliminate sexual desire and sexual behavior.3 However, the majority of older adults (defined as age ≥65) report a moderate-to-high level of sexual interest well into late life.1,3
Sexual function remains a subject often neglected in psychiatry. Sexual dysfunctions, as described in the DSM-5,4 do not include age-related changes in sexual function. In addition to physiological changes, sexual difficulties can result from relationship strain, systemic medical or psychiatric disorders, and sexual side effects of medications.
CASE REPORT
Mr. C, age 71 and married, is being treated for a major depressive episode that followed a course of shingles and persistent postherpetic neuralgia. Medications are: escitalopram, 20 mg/d; pregabalin, 150 mg/d; and ramipril, 5 mg/d. Mr. C is physically active and involved in social activities; he has no substance use history. He attends clinic visits with his wife.
Mr. C reports that despite significant improvement of his depressive and pain symptoms, he now experiences sexual difficulties, which he seems hesitant to discuss in detail. According to his wife, Mr. C appears to lack sexual desire and has difficulty initiating and maintaining an erection. She asks Mr. C’s psychiatrist whether she should stop her estrogen treatment, intended to enhance her sexual function, given that the couple is no longer engaging in sexual intercourse.
Mr. C admits to missing physical intimacy; however, he states, “If I have to make a choice between having sex with my wife and getting this depression out of my head, I’m going to pick getting rid of the depression.” Mrs. C says she is becoming dissatisfied with their marriage and the limited time she and her husband now spend together. Mr. C’s psychiatrist suggests that Mr. C and his wife undergo couples counseling.
Physiological changes with aging
In both women and men, the reproductive system undergoes age-related physiological changes.
Women. In women, the phase of decline in ovarian function and resulting decline in sex steroid production (estradiol and progesterone) is referred to as the climacteric, with menopause being determined retrospectively by the cessation of a menstrual period for 1 year.5
Menopausal symptoms typically occur between age 40 and 58; the average age of menopause is 51.6,7 Both estradiol and progesterone levels decline with menopause, and anovulation and ovarian failure ensue. A more gradual decline of female testosterone levels also occurs with aging, starting in the fourth decade of life.8
Clinical manifestations of menopause include vasomotor symptoms (ie, “hot flushes”), sleep disturbances, anxiety and depressive symptoms, decreased bone mineral density, and increased risk of cardiovascular disease.6,7 Loss of estrogen as well as continued loss of testosterone can result in dyspareunia because of atrophy and decreased vulvar and vaginal lubrication, with sexual excitement achieved less quickly, and a decreased intensity of orgasm.7
Men. Research has shown that testosterone levels are highest in men in the second and third decades, with a subsequent gradual decline.9 Older men with a low testosterone level are described as experiencing “late-onset hypogonadism,” also known by the popularized term “andropause.”10 This is attributed to decreased activity at the testicular and hypothalamic levels.10
Nonetheless, only a small fraction of older men with confirmed androgen deficiency are clinically symptomatic.11,12 Low testosterone is associated with decreased libido; it can hinder morning erections, contribute to erectile dysfunction, and result in erections that require physical stimulation.13
Notably, erectile dysfunction involves several other etiologic factors: psychiatric (eg, relationship difficulties, depression), neurogenic (eg, spinal cord injury), endocrine (eg, hyperprolactinemia), arteriogenic (eg, hypertension, type 2 diabetes mellitus), and drug-induced (eg, antidepressants, antihypertensives).14 A low testosterone level also has been associated with potential cognitive changes, decreased bone mineral density, metabolic syndrome (eg, increased risk of type 2 diabetes mellitus), and cardiovascular mortality.10
Effects on sexual activity. How much age-related physiological changes impact sexual practices in the geriatric population is uncertain. A study following 3,302 women through menopause over 6 years found some decline in sexual activity; however, this decline was not associated with increased sexual pain, decreased desire, or lack of arousal.15 Men continue to find ways to remain sexually active despite physiological changes (eg, erectile difficulties), but the etiology of sexual dysfunction in later life remains multi-modal, involving physical, psychological, and relational factors.16,17
Sexual practices in older adults
Researchers for the National Social Life, Health, and Aging Project (NSHAP) have examined sexual activities, behaviors, and problems in >3,000 older community-dwelling men and women across the United States, using information collected from in-home interviews.18 This study found that sexual activity, defined as any mutually voluntary sexual contact with another person, declines with age; however, even in the oldest age group (age 75 to 85), 39% of men and 17% of women reported being sexually active in the last 12 months. Among these persons, 54% reported sexual activity at least 2 times per month; 23% reported having sex at least once a week; and 32% reported engaging in oral sex. Partner availability predicted sexual activity.
Respondents with self-reported poor physical health were more likely to experience sexual problems (eg, difficulty with erection or lubrication, dyspareunia, and lack of pleasure). The most commonly reported reason for sexual inactivity in those with a spouse or other intimate partner was the male partner’s poor physical health.18
A longitudinal study, part of the Women’s Healthy Ageing Project, examined changes in sexual function at late menopause compared with early menopause. Although the researchers also found an age-related decrease in sexual activity, 50% of their late-menopause respondents (mean age, 70; range, 64 to 77) still reported sexual activity in the previous month, with 35% of this subgroup reporting sexual activity at least once a week; 83% reported sexual thoughts or fantasies.19 Availability of a partner, absence of a history of depression, moderate (compared with no) alcohol consumption, and better cognitive function were significantly associated with a higher level of sexual activity.19
In the Successful Aging Evaluation study, conducted in San Diego County, California, community-dwelling older partnered adults age 50 to 99 (mean age, 75) were surveyed about their sexual health after a cognitive screen by telephone20; rating scales for depression, anxiety, and physical function also were included. Results included 41% of men and 35% of women reporting sexual activity at least once a week, and only 21% of men and 24% of women reporting no sexual activity in the previous year. Depressive symptoms were most highly correlated with lack of sexual activity.20
Overall, these studies reveal that positive physical and mental health, access to a healthy partner, and a positive attitude toward sex are correlated with sexual activity in later life, whereas barriers to sexual activity include lack of a healthy sexual partner, depression, and chronic systemic medical illnesses, such as coronary artery disease or type 2 diabetes mellitus.13,17,21-24 Sexual activity and satisfaction have been positively associated with perceived general well-being and self-esteem.25,26 Conversely, sexual difficulties secondary to disease can be a source of distress for couples.27
Possibly overlooked? It is important to note that sexuality itself is a subjective area. Psychological intimacy is a universal phenomenon, and its physical expression may evolve as couples accommodate to age-related bodily changes. Means of achieving physical closeness, other than intercourse (eg, intimate touching, hand holding, kissing, or even acts of caretaking), may not be adequately captured in studies that look specifically at sexual activity.
Taking a sexual history in a geriatric patient
Because sexuality can be an uncomfortable topic for geriatric patients to discuss, sexual problems in this population often go unrecognized. It has been suggested that psychiatrists are more likely to inquire about sexual activity in middle-aged patients than geriatric patients with the same psychiatric presentation—perhaps illustrating a bias against taking a sexual history from a geriatric patient.28 However, because many older patients can experience depression or anxiety disorders in relation to normal sexual changes or sexual dysfunction within the context of their intimate relationships, it is essential to bring these issues to light.
Although a sexual history may not be the focus of a first clinical encounter, consider making such an assessment at a relatively early stage of patient care. The importance of such dialogue is 2-fold:
• It demonstrates to the patient that talking about sexuality in a respectful and empathic manner is appropriate and can encourage patients to communicate more effectively about sexuality with clinicians and with sexual partners.
• It helps elicit medical information needed to make an accurate diagnosis and provide adequate management.
How to begin. As a starting point to taking a sexual history, an open-ended invitation for the geriatric patient to share information may be best, such as “What would you like to tell me about your sexual life?” See further suggestions (Table 1) and examples of more detailed questions to ask once a dialogue has been initiated (Table 2). Additional factors that may contribute to sexual dysfunction are presented in Table 3.1,27,29,30
CASE CONTINUED
In Mr. C’s case, an assessment of his sexual history, including risk factors for sexual dysfunction, is completed. Results from laboratory investigations, including a total testosterone level, are within normal limits.
Mr. C asks about using medications with fewer sexual side effects (he has been taking 3 medications that can contribute to sexual dysfunction). A gradual cross-taper of escitalopram, 20 mg/d, to mirtazapine, 45 mg/d, is implemented, along with tapering pregabalin to 50 mg/d.
Mr. C’s psychiatric and pain symptom improvement is maintained. He notices a boost in his sexual desire but has minimal improvement in erectile dysfunction. He is encouraged to speak with his primary care physician about an antihypertensive agent with less impact on sexual function, as well as therapeutic agents for erectile dysfunction; these, he declines.
At a subsequent visit, Mr. C reports feeling less apprehension about sexual performance. He is now willing to consider further medication options with his primary care physician, and agrees to a recommendation for couples psychotherapy.
As illustrated in Mr. C’s case, the recommended sexual assessment and management strategies to consider at a minimum in psychiatric practice are listed in Table 4.
STI risk in geriatric patients
The risk of sexually transmitted infections (STIs), including human immunodeficiency virus (HIV), often is overlooked in sexually active older adults. Although STIs are more common among younger adults, there is recent evidence of increased incidence in the geriatric population31 (with the highest risk of incident HIV and some STIs in older men who have sex with men32). These increased rates can be explained, at least in part, by:
• older men being less likely to use a condom during sexual activity
• promotion of viral entry in older women through a drier, thinner vaginal wall
• increased longevity of HIV-positive persons.31
Routine STI screening is not warranted in all older adults, but education and prevention strategies in sexually active seniors who are at greater risk of STIs are recommended. Particularly, clinicians should seek opportunities to discuss risk factors and safe sex practices (eg, using condoms, limiting number of sexual partners, practicing good hygiene, engaging in preventive care), and provide behavioral counseling where appropriate.31,33
Additional considerations in geriatric sexuality
Because psychiatric and systemic medical conditions can hinder sexual function, it is essential to identify and manage these conditions. Several neuropsychiatric disorders, including mood and neurocognitive disorders, can not only cause sexual dysfunction, but also can raise ethical dilemmas for clinicians, such as reduced decisional capacity in cognitively impaired patients to consent to sexual activity.1,34
In some patients, psychological, environmental, and pharmacological treatment options may help. A phosphodiesterase type 5 inhibitor for erectile dysfunction can be prescribed by the primary care physician, a psychiatrist, or another specialist, depending on the physician’s expertise and comfort level.
Sequencing of sexual dysfunction. Notably, there is a common paradox in mood disorders. Decreased sexual interest or performance may represent an aspect of anhedonia associated with depression, whereas sexual dysfunction could also result from medication use (particularly that of serotonergic antidepressants, such as selective serotonin reuptake inhibitors and serotonin-norepinephrine inhibitors), even as other depressive symptoms improve. Therefore, it is critical to analyze sequencing of sexual dysfunction—as part of the presenting mood symptoms or dysfunction induced by antidepressant treatment.
Geriatric sexuality in the digital age. Because older adults represent a rapidly growing segment of digital device users,35 Internet use is likely to play a role in the future of sexuality and “digital intimacy,” in that older adults can engage in online sexual activities. The Internet also can be a tool to access medical education.
Related Resources
• Burghardt KJ, Gardner KN. Sildenafil for SSRI-induced sexual dysfunction. Current Psychiatry. 2013;12(4):29-32,A.
• Maciel M, Laganà L. Older women’s sexual desire problems: biopsychosocial factors impacting them and barriers to their clinical assessment [published online January 5, 2014]. Biomed Res Int. 2014;2014:107217. doi: 10.1155/2014/107217.
Drug Brand Names
Bupropion • Wellbutrin, Zyban Mirtazapine • Remeron
Carbamazepine • Tegretol Oxcarbazepine • Trileptal
Clonidine • Catapres Phenobarbital • Luminal
Donepezil • Aricept Phenytoin • Dilantin
Escitalopram • Lexapro Pregabalin • Lyrica
Gabapentin • Neurontin Ramipril • Altace
Lamotrigine • Lamictal Rivastigmine • Exelon
Lithium • Eskalith, Lithobid Trazodone • Desyrel
Memantine • Namenda Valproic acid • Depakote
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Jagus CE, Benbow SM. Sexuality in older men with mental health problems. Sex Relation Ther. 2002;17(3):271-279.
2. Hillman JL. Clinical perspectives on elderly sexuality. New York, NY: Springer; 2000.
3. DeLamater JD, Sill M. Sexual desire in later life. J Sex Res. 2005;42(2):138-149.
4. Diagnostic and statistical manual of mental disorders. 5th ed. Washington, DC: American Psychiatric Association; 2013.
5. Laufer LR, Gambone JC. Climacteric: menopause and peri-and postmenopause. In: Hacker NF, Gambone JC, Hobel CJ. Hacker and Moore’s essentials of obstetrics and gynecology. 5th ed. Philadelphia, PA: Saunders/Elsevier; 2010:379-385.
6. Wilson MM. Menopause. Clin Geriatr Med. 2003;19(3): 483-506.
7. Reid R, Abramson BL, Blake J, et al. Managing menopause. J Obstet Gynaecol Can. 2014;36(9):830-838.
8. Horstman AM, Dillon EL, Urban RJ, et al. The role of androgens and estrogens on healthy aging and longevity. J Gerontol A Biol Sci Med Sci. 2012;67(11):1140-1152.
9. Wu FC, Tajar A, Pye SR, et al. Hypothalamic-pituitary-testicular axis disruptions in older men are differentially linked to age and modifiable risk factors: the European Male Aging Study. J Clin Endocrinol Metab. 2008;93(7):2737-2745.
10. Basaria S. Reproductive aging in men. Endocrinol Metab Clin North Am. 2013;42(2):255-270.
11. Wu FC, Tajar A, Beynon JM, et al. Identification of late-onset hypogonadism in middle-aged and elderly men. N Engl J Med. 2010;363(2):123-135.
12. Araujo AB, Esche GR, Kupelian V, et al. Prevalence of symptomatic androgen deficiency in men. J Clin Endocrinol Metab. 2007;92(11):4241-4247.
13. Lochlainn MN, Kenny RA. Sexual activity and aging. J Am Med Dir Assoc. 2013;14(8):565-572.
14. McMahon CG. Erectile dysfunction. Intern Med J. 2014;44(1):18-26.
15. Avis NE, Brockwell S, Randolph JF Jr, et al. Longitudinal changes in sexual functioning as women transition through menopause: results from the Study of Women’s Health Across the Nation. Menopause. 2009;16(3):442-452.
16. Perelman M, Shabsigh R, Seftel A, et al. Attitudes of men with erectile dysfunction: a cross-national survey. J Sex Med. 2005;2(3):397-406.
17. Corona G, Rastrelli G, Maseroli E, et al. Sexual function of the ageing male. Best Pract Res Clin Endocrinol Metab. 2013;27(4):581-601.
18. Lindau ST, Schumm LP, Laumann EO, et al. A study of sexuality and health among older adults in the United States. N Engl J Med. 2007;357(8):762-774.
19. Lonnèe-Hoffmann RA, Dennerstein L, Lehert P, et al. Sexual function in the late postmenopause: a decade of follow-up in a population-based cohort of Australian women. J Sex Med. 2014;11(8):2029-2038.
20. Wang V, Depp CA, Ceglowski J, et al. Sexual health and function in later life: a population-based study of 606 older adults with a partner. Am J Geriatr Psychiatry. 2015;23(3):227-233.
21. Garrett D. Psychosocial barriers to sexual intimacy for older people. Br J Nurs. 2014;23(6):327-331.
22. DeLamater J, Karraker A. Sexual functioning in older adults. Curr Psychiatry Rep. 2009;11(1):6-11.
23. DeLamater J. Sexual expression in later life: a review and synthesis. J Sex Res. 2012;49(2-3):125-141.
24. Inelmen EM, Sergi G, Girardi A, et al. The importance of sexual health in the elderly: breaking down barriers and taboos. Aging Clin Exp Res. 2012;24(suppl 3):31-34.
25. Choi KB, Jang SH, Lee MY, et al. Sexual life and self-esteem in married elderly. Arch Gerontol Geriatr. 2011;53(1):e17-e20.
26. Davison SL, Bell RJ, LaChina M, et al. The relationship between self-reported sexual satisfaction and general well-being in women. J Sex Med. 2009;6(10):2690-2697.
27. Morley JE, Tariq SH. Sexuality and disease. Clin Geriatr Med. 2003;19(3):563-573.
28. Bouman WP, Arcelus J. Are psychiatrists guilty of “ageism” when it comes to taking a sexual history? Int J Geriatr Psychiatry. 2001;16(1):27-31.
29. La Torre A, Giupponi G, Duffy DM, et al. Sexual dysfunction related to psychotropic drugs: a critical review. Part III: mood stabilizers and anxiolytic drugs. Pharmacopsychiatry. 2014;47(1):1-6.
30. Tucker I. Management of inappropriate sexual behaviors in dementia: a literature review. Int Psychogeriatr. 2010; 22(5):683-692.
31. Imparato T, Sanders D. STD prevalence demands clinical awareness. Aging Well. 2012;5(1):14.
32. Poynten IM, Grulich AE, Templeton DJ. Sexually transmitted infections in older populations. Curr Opin Infect Dis. 2013;26(1):80-85.
33. Talashek ML, Tichy AM, Epping H. Sexually transmitted diseases in the elderly—issues and recommendations. J Gerontol Nurs. 1990;16(4):33-40.
34. Benbow SM, Jagus CE. Sexuality in older women with mental health problems. Sex Relation Ther. 2002;17(3):261-270.
35. Veenhof B, Timusk P. Online activities of Canadian boomers and seniors. http://www.statcan.gc.ca/pub/ 11-008-x/2009002/article/10910-eng.htm#tphp. Accessed March 26, 2015.
1. Jagus CE, Benbow SM. Sexuality in older men with mental health problems. Sex Relation Ther. 2002;17(3):271-279.
2. Hillman JL. Clinical perspectives on elderly sexuality. New York, NY: Springer; 2000.
3. DeLamater JD, Sill M. Sexual desire in later life. J Sex Res. 2005;42(2):138-149.
4. Diagnostic and statistical manual of mental disorders. 5th ed. Washington, DC: American Psychiatric Association; 2013.
5. Laufer LR, Gambone JC. Climacteric: menopause and peri-and postmenopause. In: Hacker NF, Gambone JC, Hobel CJ. Hacker and Moore’s essentials of obstetrics and gynecology. 5th ed. Philadelphia, PA: Saunders/Elsevier; 2010:379-385.
6. Wilson MM. Menopause. Clin Geriatr Med. 2003;19(3): 483-506.
7. Reid R, Abramson BL, Blake J, et al. Managing menopause. J Obstet Gynaecol Can. 2014;36(9):830-838.
8. Horstman AM, Dillon EL, Urban RJ, et al. The role of androgens and estrogens on healthy aging and longevity. J Gerontol A Biol Sci Med Sci. 2012;67(11):1140-1152.
9. Wu FC, Tajar A, Pye SR, et al. Hypothalamic-pituitary-testicular axis disruptions in older men are differentially linked to age and modifiable risk factors: the European Male Aging Study. J Clin Endocrinol Metab. 2008;93(7):2737-2745.
10. Basaria S. Reproductive aging in men. Endocrinol Metab Clin North Am. 2013;42(2):255-270.
11. Wu FC, Tajar A, Beynon JM, et al. Identification of late-onset hypogonadism in middle-aged and elderly men. N Engl J Med. 2010;363(2):123-135.
12. Araujo AB, Esche GR, Kupelian V, et al. Prevalence of symptomatic androgen deficiency in men. J Clin Endocrinol Metab. 2007;92(11):4241-4247.
13. Lochlainn MN, Kenny RA. Sexual activity and aging. J Am Med Dir Assoc. 2013;14(8):565-572.
14. McMahon CG. Erectile dysfunction. Intern Med J. 2014;44(1):18-26.
15. Avis NE, Brockwell S, Randolph JF Jr, et al. Longitudinal changes in sexual functioning as women transition through menopause: results from the Study of Women’s Health Across the Nation. Menopause. 2009;16(3):442-452.
16. Perelman M, Shabsigh R, Seftel A, et al. Attitudes of men with erectile dysfunction: a cross-national survey. J Sex Med. 2005;2(3):397-406.
17. Corona G, Rastrelli G, Maseroli E, et al. Sexual function of the ageing male. Best Pract Res Clin Endocrinol Metab. 2013;27(4):581-601.
18. Lindau ST, Schumm LP, Laumann EO, et al. A study of sexuality and health among older adults in the United States. N Engl J Med. 2007;357(8):762-774.
19. Lonnèe-Hoffmann RA, Dennerstein L, Lehert P, et al. Sexual function in the late postmenopause: a decade of follow-up in a population-based cohort of Australian women. J Sex Med. 2014;11(8):2029-2038.
20. Wang V, Depp CA, Ceglowski J, et al. Sexual health and function in later life: a population-based study of 606 older adults with a partner. Am J Geriatr Psychiatry. 2015;23(3):227-233.
21. Garrett D. Psychosocial barriers to sexual intimacy for older people. Br J Nurs. 2014;23(6):327-331.
22. DeLamater J, Karraker A. Sexual functioning in older adults. Curr Psychiatry Rep. 2009;11(1):6-11.
23. DeLamater J. Sexual expression in later life: a review and synthesis. J Sex Res. 2012;49(2-3):125-141.
24. Inelmen EM, Sergi G, Girardi A, et al. The importance of sexual health in the elderly: breaking down barriers and taboos. Aging Clin Exp Res. 2012;24(suppl 3):31-34.
25. Choi KB, Jang SH, Lee MY, et al. Sexual life and self-esteem in married elderly. Arch Gerontol Geriatr. 2011;53(1):e17-e20.
26. Davison SL, Bell RJ, LaChina M, et al. The relationship between self-reported sexual satisfaction and general well-being in women. J Sex Med. 2009;6(10):2690-2697.
27. Morley JE, Tariq SH. Sexuality and disease. Clin Geriatr Med. 2003;19(3):563-573.
28. Bouman WP, Arcelus J. Are psychiatrists guilty of “ageism” when it comes to taking a sexual history? Int J Geriatr Psychiatry. 2001;16(1):27-31.
29. La Torre A, Giupponi G, Duffy DM, et al. Sexual dysfunction related to psychotropic drugs: a critical review. Part III: mood stabilizers and anxiolytic drugs. Pharmacopsychiatry. 2014;47(1):1-6.
30. Tucker I. Management of inappropriate sexual behaviors in dementia: a literature review. Int Psychogeriatr. 2010; 22(5):683-692.
31. Imparato T, Sanders D. STD prevalence demands clinical awareness. Aging Well. 2012;5(1):14.
32. Poynten IM, Grulich AE, Templeton DJ. Sexually transmitted infections in older populations. Curr Opin Infect Dis. 2013;26(1):80-85.
33. Talashek ML, Tichy AM, Epping H. Sexually transmitted diseases in the elderly—issues and recommendations. J Gerontol Nurs. 1990;16(4):33-40.
34. Benbow SM, Jagus CE. Sexuality in older women with mental health problems. Sex Relation Ther. 2002;17(3):261-270.
35. Veenhof B, Timusk P. Online activities of Canadian boomers and seniors. http://www.statcan.gc.ca/pub/ 11-008-x/2009002/article/10910-eng.htm#tphp. Accessed March 26, 2015.