User login
Evaluating the Implementation of 60-minute Iron Dextran Infusions at a Rural Health Center
Background
Due to risk for infusion-related reactions (IRR), administration of iron dextran requires an initial test dose with an extended monitoring period and subsequent doses given as a slow infusion over 2-3 hours. Safe use of a 60-minute iron dextran infusion protocol has been demonstrated previously at fully staffed academic teaching institutions. This study sought to determine the impact on patient safety and infusion clinic efficiency after implementing a 60-minute iron dextran administration protocol at a small, rural facility utilizing a decentralized clinical model.
Methods
This single-site, prospective, interventional study was conducted at a rural level 1C Veterans Affairs secondary care facility. The Hematology/Oncology clinic staffing includes one onsite clinical pharmacy practitioner (CPP) and advanced practice nurse. Remote providers complete patient encounters through video and telehealth modalities. A 60-minute iron dextran infusion service line agreement was designed by the Hematology/Oncology CPP and approved by the facility prior to data collection. The protocol included administration of a test dose and 15-minute monitoring period for treatment naïve patients. Pre-medications were allowed at the discretion of the ordering providers. All patients who received iron dextran between May 31, 2024 and April 14, 2025 per protocol were included in data analysis and results were stratified by treatment naïve and pre-treated patients. Outcomes included the proportion of patients experiencing any grade of IRR based on the Common Criteria for Adverse Events Version 5.0, and the average duration of administration. Descriptive statistics were used for safety and efficiency outcomes.
Results
Eighty patients received 103 iron dextran infusions and were included for analysis. Pre-medications were administered for 16 of the 103 (15.5%) included infusions. Two patients experienced grade 1 IRR (nausea) on 4 occasions (3.8%) which quickly resolved with intravenous ondansetron, and full iron dextran doses were received. The mean infusion time was 94 minutes in the treatment naïve cohort vs 71 minutes in the pre-treated cohort.
Conclusions
This study suggests a Hematology/ Oncology CPP developed iron dextran 60-minute infusion protocol may be safely and efficiently administered for qualifying patients in a decentralized, rural healthcare setting.
Background
Due to risk for infusion-related reactions (IRR), administration of iron dextran requires an initial test dose with an extended monitoring period and subsequent doses given as a slow infusion over 2-3 hours. Safe use of a 60-minute iron dextran infusion protocol has been demonstrated previously at fully staffed academic teaching institutions. This study sought to determine the impact on patient safety and infusion clinic efficiency after implementing a 60-minute iron dextran administration protocol at a small, rural facility utilizing a decentralized clinical model.
Methods
This single-site, prospective, interventional study was conducted at a rural level 1C Veterans Affairs secondary care facility. The Hematology/Oncology clinic staffing includes one onsite clinical pharmacy practitioner (CPP) and advanced practice nurse. Remote providers complete patient encounters through video and telehealth modalities. A 60-minute iron dextran infusion service line agreement was designed by the Hematology/Oncology CPP and approved by the facility prior to data collection. The protocol included administration of a test dose and 15-minute monitoring period for treatment naïve patients. Pre-medications were allowed at the discretion of the ordering providers. All patients who received iron dextran between May 31, 2024 and April 14, 2025 per protocol were included in data analysis and results were stratified by treatment naïve and pre-treated patients. Outcomes included the proportion of patients experiencing any grade of IRR based on the Common Criteria for Adverse Events Version 5.0, and the average duration of administration. Descriptive statistics were used for safety and efficiency outcomes.
Results
Eighty patients received 103 iron dextran infusions and were included for analysis. Pre-medications were administered for 16 of the 103 (15.5%) included infusions. Two patients experienced grade 1 IRR (nausea) on 4 occasions (3.8%) which quickly resolved with intravenous ondansetron, and full iron dextran doses were received. The mean infusion time was 94 minutes in the treatment naïve cohort vs 71 minutes in the pre-treated cohort.
Conclusions
This study suggests a Hematology/ Oncology CPP developed iron dextran 60-minute infusion protocol may be safely and efficiently administered for qualifying patients in a decentralized, rural healthcare setting.
Background
Due to risk for infusion-related reactions (IRR), administration of iron dextran requires an initial test dose with an extended monitoring period and subsequent doses given as a slow infusion over 2-3 hours. Safe use of a 60-minute iron dextran infusion protocol has been demonstrated previously at fully staffed academic teaching institutions. This study sought to determine the impact on patient safety and infusion clinic efficiency after implementing a 60-minute iron dextran administration protocol at a small, rural facility utilizing a decentralized clinical model.
Methods
This single-site, prospective, interventional study was conducted at a rural level 1C Veterans Affairs secondary care facility. The Hematology/Oncology clinic staffing includes one onsite clinical pharmacy practitioner (CPP) and advanced practice nurse. Remote providers complete patient encounters through video and telehealth modalities. A 60-minute iron dextran infusion service line agreement was designed by the Hematology/Oncology CPP and approved by the facility prior to data collection. The protocol included administration of a test dose and 15-minute monitoring period for treatment naïve patients. Pre-medications were allowed at the discretion of the ordering providers. All patients who received iron dextran between May 31, 2024 and April 14, 2025 per protocol were included in data analysis and results were stratified by treatment naïve and pre-treated patients. Outcomes included the proportion of patients experiencing any grade of IRR based on the Common Criteria for Adverse Events Version 5.0, and the average duration of administration. Descriptive statistics were used for safety and efficiency outcomes.
Results
Eighty patients received 103 iron dextran infusions and were included for analysis. Pre-medications were administered for 16 of the 103 (15.5%) included infusions. Two patients experienced grade 1 IRR (nausea) on 4 occasions (3.8%) which quickly resolved with intravenous ondansetron, and full iron dextran doses were received. The mean infusion time was 94 minutes in the treatment naïve cohort vs 71 minutes in the pre-treated cohort.
Conclusions
This study suggests a Hematology/ Oncology CPP developed iron dextran 60-minute infusion protocol may be safely and efficiently administered for qualifying patients in a decentralized, rural healthcare setting.
Implementation of a VHA Virtual Oncology Training Pilot Program for Clinical Pharmacists
Purpose/Background
Oncology clinical pharmacist practitioners (CPP) play a critical role in optimizing drug therapy, managing side effects, and ensuring medication adherence. As a specialized clinical area, specific training is needed to ensure quality of care. Oncology pharmacy training programs are commercially available but pose a financial burden and are not specific to the Veterans Health Administration (VHA). A comprehensive, virtual Oncology Bootcamp series was implemented to upskill new oncology pharmacists (or pharmacists seeking to further their understanding of oncology practice), with didactic materials and clinical tools to enhance and standardize quality care delivery.
Methods
This program was comprised of an online platform of 23 one hour-long continuing education accredited sessions, delivered by leading subject matter experts. Pharmacists from two Veteran Integrated Service Networks (VISNs) were invited for the first year of the bootcamp. The curriculum encompassed fundamentals of oncology practice, patient care assessment, chemotherapy protocol review, practice management, and supportive care. Participants also received in-depth training on managing various cancer types, including but not limited to prostate, lung, gastrointestinal and hematologic malignancies. VHA specific information, including utilization of Oncology Clinical Pathways to promote standardized care was included where applicable. The interactive nature of the virtual sessions provided opportunities for real-time discussion and immediate feedback. To measure the impact of this program, a pre and post program evaluation of participants was conducted.
Results
Over the course of the program, more than 40 pharmacists across two VISNs participated in the bootcamp series. Results of the program evaluation showed an increase in self-reported comfort and skill levels in all criteria that were assessed (oncology pharmacotherapy, solid tumor malignancies, hematologic malignancies and oral anti-cancer therapy management). Additionally, 85% of respondents stated the series met their overall goals and over 90% of respondents stated they were either satisfied or very satisfied with the content, speakers and organization of the course.
Implications/Significance
This initiative has established the viability and significance of a highly accessible, VHA pathway specific and Veteran centric platform for oncology pharmacy professional development. Future directions for the program include a broader nationwide audience, increased content coverage and self-paced learning options.
Purpose/Background
Oncology clinical pharmacist practitioners (CPP) play a critical role in optimizing drug therapy, managing side effects, and ensuring medication adherence. As a specialized clinical area, specific training is needed to ensure quality of care. Oncology pharmacy training programs are commercially available but pose a financial burden and are not specific to the Veterans Health Administration (VHA). A comprehensive, virtual Oncology Bootcamp series was implemented to upskill new oncology pharmacists (or pharmacists seeking to further their understanding of oncology practice), with didactic materials and clinical tools to enhance and standardize quality care delivery.
Methods
This program was comprised of an online platform of 23 one hour-long continuing education accredited sessions, delivered by leading subject matter experts. Pharmacists from two Veteran Integrated Service Networks (VISNs) were invited for the first year of the bootcamp. The curriculum encompassed fundamentals of oncology practice, patient care assessment, chemotherapy protocol review, practice management, and supportive care. Participants also received in-depth training on managing various cancer types, including but not limited to prostate, lung, gastrointestinal and hematologic malignancies. VHA specific information, including utilization of Oncology Clinical Pathways to promote standardized care was included where applicable. The interactive nature of the virtual sessions provided opportunities for real-time discussion and immediate feedback. To measure the impact of this program, a pre and post program evaluation of participants was conducted.
Results
Over the course of the program, more than 40 pharmacists across two VISNs participated in the bootcamp series. Results of the program evaluation showed an increase in self-reported comfort and skill levels in all criteria that were assessed (oncology pharmacotherapy, solid tumor malignancies, hematologic malignancies and oral anti-cancer therapy management). Additionally, 85% of respondents stated the series met their overall goals and over 90% of respondents stated they were either satisfied or very satisfied with the content, speakers and organization of the course.
Implications/Significance
This initiative has established the viability and significance of a highly accessible, VHA pathway specific and Veteran centric platform for oncology pharmacy professional development. Future directions for the program include a broader nationwide audience, increased content coverage and self-paced learning options.
Purpose/Background
Oncology clinical pharmacist practitioners (CPP) play a critical role in optimizing drug therapy, managing side effects, and ensuring medication adherence. As a specialized clinical area, specific training is needed to ensure quality of care. Oncology pharmacy training programs are commercially available but pose a financial burden and are not specific to the Veterans Health Administration (VHA). A comprehensive, virtual Oncology Bootcamp series was implemented to upskill new oncology pharmacists (or pharmacists seeking to further their understanding of oncology practice), with didactic materials and clinical tools to enhance and standardize quality care delivery.
Methods
This program was comprised of an online platform of 23 one hour-long continuing education accredited sessions, delivered by leading subject matter experts. Pharmacists from two Veteran Integrated Service Networks (VISNs) were invited for the first year of the bootcamp. The curriculum encompassed fundamentals of oncology practice, patient care assessment, chemotherapy protocol review, practice management, and supportive care. Participants also received in-depth training on managing various cancer types, including but not limited to prostate, lung, gastrointestinal and hematologic malignancies. VHA specific information, including utilization of Oncology Clinical Pathways to promote standardized care was included where applicable. The interactive nature of the virtual sessions provided opportunities for real-time discussion and immediate feedback. To measure the impact of this program, a pre and post program evaluation of participants was conducted.
Results
Over the course of the program, more than 40 pharmacists across two VISNs participated in the bootcamp series. Results of the program evaluation showed an increase in self-reported comfort and skill levels in all criteria that were assessed (oncology pharmacotherapy, solid tumor malignancies, hematologic malignancies and oral anti-cancer therapy management). Additionally, 85% of respondents stated the series met their overall goals and over 90% of respondents stated they were either satisfied or very satisfied with the content, speakers and organization of the course.
Implications/Significance
This initiative has established the viability and significance of a highly accessible, VHA pathway specific and Veteran centric platform for oncology pharmacy professional development. Future directions for the program include a broader nationwide audience, increased content coverage and self-paced learning options.
Daratumumab and Darbepoetin for Refractory Warm Autoimmune Hemolytic Anemia: A Novel Duo for a Tough Case
Background
Warm autoimmune hemolytic anemia (wAIHA) is traditionally treated with immunosuppresimmunosuppression, and management of refractory disease is often a challenge. The anti-CD38 antibody daratumumab is emerging as a promising treatment for refractory wAIHA, as it targets autoantibody-producing plasma cells. Here, we present the first reported case of daratumumab used in conjunction with an erythropoiesisstimulating agent (ESA) to salvage refractory wAIHA in a patient with AIDS and bone marrow suppression.
Case Presentation
A middle aged man with HIV (undetectable viral load on antiretroviral treatment but CD4 persistently < 200, requiring chronic antimicrobial prophylaxis) was diagnosed with classic wAIHA in late 2021. The disease initially responded to corticosteroids, but relapsed repeatedly and eventually required IVIG, rituximab, danazol, and three immunosuppressive agents, none of which induced remission. Hemolysis worsened by fall 2024, with hemoglobin 5-6 g/dL despite high-dose corticosteroids and IVIG. Bone marrow biopsy was unrevealing, and he underwent splenectomy. However, recovery was complicated by cutaneous nocardiosis, iron overload, liver injury, and continued hemolysis. Eventually, reticulocytosis also ceased, and hemoglobin declined to 4-5 g/dL. Due to failure of standard therapies and to minimize further immunosuppression, weekly daratumumab injections were initiated, with weekly darbepoetin injections added to aid in compensatory hematopoiesis. With this combination, hemolysis indices improved, reticulocytosis picked up, and hemoglobin increased to 8-9 g/dL. However, the patient continued to struggle with infections, and he succumbed to drug-resistant bacterial sepsis in spring 2025.
Discussion
The patient had very complicated chronic and acute comorbidities, and some simplification was required in order to provide this summary. However, we hope this case adds to the literature on daratumumab as an effective new agent in refractory wAIHA, and also present a novel duo of therapies for patients who may struggle with bone marrow suppression in addition to autoimmune hemolysis. To our knowledge, this is the first reported case of the combination used in this manner.
Conclusions
Daratumumab is an effective and less immunosuppressive alternative for the treatment of heavily pretreated refractory wAIHA. Its combined use with ESA in patients with inadequate reticulocytosis should be studied further to clarify the efficacy and safety in this setting.
Background
Warm autoimmune hemolytic anemia (wAIHA) is traditionally treated with immunosuppresimmunosuppression, and management of refractory disease is often a challenge. The anti-CD38 antibody daratumumab is emerging as a promising treatment for refractory wAIHA, as it targets autoantibody-producing plasma cells. Here, we present the first reported case of daratumumab used in conjunction with an erythropoiesisstimulating agent (ESA) to salvage refractory wAIHA in a patient with AIDS and bone marrow suppression.
Case Presentation
A middle aged man with HIV (undetectable viral load on antiretroviral treatment but CD4 persistently < 200, requiring chronic antimicrobial prophylaxis) was diagnosed with classic wAIHA in late 2021. The disease initially responded to corticosteroids, but relapsed repeatedly and eventually required IVIG, rituximab, danazol, and three immunosuppressive agents, none of which induced remission. Hemolysis worsened by fall 2024, with hemoglobin 5-6 g/dL despite high-dose corticosteroids and IVIG. Bone marrow biopsy was unrevealing, and he underwent splenectomy. However, recovery was complicated by cutaneous nocardiosis, iron overload, liver injury, and continued hemolysis. Eventually, reticulocytosis also ceased, and hemoglobin declined to 4-5 g/dL. Due to failure of standard therapies and to minimize further immunosuppression, weekly daratumumab injections were initiated, with weekly darbepoetin injections added to aid in compensatory hematopoiesis. With this combination, hemolysis indices improved, reticulocytosis picked up, and hemoglobin increased to 8-9 g/dL. However, the patient continued to struggle with infections, and he succumbed to drug-resistant bacterial sepsis in spring 2025.
Discussion
The patient had very complicated chronic and acute comorbidities, and some simplification was required in order to provide this summary. However, we hope this case adds to the literature on daratumumab as an effective new agent in refractory wAIHA, and also present a novel duo of therapies for patients who may struggle with bone marrow suppression in addition to autoimmune hemolysis. To our knowledge, this is the first reported case of the combination used in this manner.
Conclusions
Daratumumab is an effective and less immunosuppressive alternative for the treatment of heavily pretreated refractory wAIHA. Its combined use with ESA in patients with inadequate reticulocytosis should be studied further to clarify the efficacy and safety in this setting.
Background
Warm autoimmune hemolytic anemia (wAIHA) is traditionally treated with immunosuppresimmunosuppression, and management of refractory disease is often a challenge. The anti-CD38 antibody daratumumab is emerging as a promising treatment for refractory wAIHA, as it targets autoantibody-producing plasma cells. Here, we present the first reported case of daratumumab used in conjunction with an erythropoiesisstimulating agent (ESA) to salvage refractory wAIHA in a patient with AIDS and bone marrow suppression.
Case Presentation
A middle aged man with HIV (undetectable viral load on antiretroviral treatment but CD4 persistently < 200, requiring chronic antimicrobial prophylaxis) was diagnosed with classic wAIHA in late 2021. The disease initially responded to corticosteroids, but relapsed repeatedly and eventually required IVIG, rituximab, danazol, and three immunosuppressive agents, none of which induced remission. Hemolysis worsened by fall 2024, with hemoglobin 5-6 g/dL despite high-dose corticosteroids and IVIG. Bone marrow biopsy was unrevealing, and he underwent splenectomy. However, recovery was complicated by cutaneous nocardiosis, iron overload, liver injury, and continued hemolysis. Eventually, reticulocytosis also ceased, and hemoglobin declined to 4-5 g/dL. Due to failure of standard therapies and to minimize further immunosuppression, weekly daratumumab injections were initiated, with weekly darbepoetin injections added to aid in compensatory hematopoiesis. With this combination, hemolysis indices improved, reticulocytosis picked up, and hemoglobin increased to 8-9 g/dL. However, the patient continued to struggle with infections, and he succumbed to drug-resistant bacterial sepsis in spring 2025.
Discussion
The patient had very complicated chronic and acute comorbidities, and some simplification was required in order to provide this summary. However, we hope this case adds to the literature on daratumumab as an effective new agent in refractory wAIHA, and also present a novel duo of therapies for patients who may struggle with bone marrow suppression in addition to autoimmune hemolysis. To our knowledge, this is the first reported case of the combination used in this manner.
Conclusions
Daratumumab is an effective and less immunosuppressive alternative for the treatment of heavily pretreated refractory wAIHA. Its combined use with ESA in patients with inadequate reticulocytosis should be studied further to clarify the efficacy and safety in this setting.
Successful Targeted Therapy with Alectinib in ALK-Positive Metastatic Pancreatic Cancer
Background
Pancreatic cancer has one of the highest mortality rates due to its typical late-stage diagnosis and subsequent limited surgical options. However, recent advances in molecular profiling offer hope for targeted therapies. We present a case of locally advanced pancreatic adenocarcinoma which progressed despite surgery and chemotherapy yet showed a positive respond to Alectinib.
Case Description
A 79-year-old male with medical history of tobacco use and ulcerative colitis presented to the clinic with 15lb unintentional weight loss over the past few months in 04/2021. Computed tomography (CT) showed dilated common bile duct due to 2.2 x 1.9 x 1.7 cm mass with no metastatic disease. Biopsy was consistent with pancreatic adenocarcinoma and patient completed 6 cycles of dose-reduced neoadjuvant gemcitabine and paclitaxel in late 2021 due to his severe neuropathy and ECOG. Subsequent CT and PET-CT showed stable disease prior to undergoing pylorus-sparing pancreatoduodenectomy and cholecystectomy with portal vein resection in 05/2022 with surgical pathology grading yPT4N2cM0. The follow- up PET scan in 09/2022 revealed new pulmonary and liver metastases, along with increased uptake in the pancreatic region, suggesting recurrent disease. Next generation sequencing (NGS) identified an ELM4-ALK chromosomal rearrangement on the surgical pathology. Given the patient’s cancer progression and concerns about chemotherapy tolerance, Alectinib, a second-generation ALK inhibitor more commonly used in lung cancer, was considered as a treatment option. Patient began Alectinib 10/2022 with no significant side effects and PET scan on 03/2023 and 06/2023 showing resolution of his lung nodules and liver lesions. Patient remained on Alectinib until he transitioned to hospice after an ischemic stroke in 03/2024.
Discussion
Pancreatic cancer urgently requires novel therapies as about 25% of patients harbor actionable molecular alterations that have led to the success of targeted therapies. ALK fusion genes are identified in multiple cancers, but the prevalence is only 0.16% in pancreatic ductal adenocarcinoma. Alectinib provided an extended progression free survival compared with standard chemotherapy in our patient. ALK inhibitors may demonstrate a remarkable response in metastatic pancreatic cancer even in poor candidates for standard chemotherapy highlighting the emphasis of NGS and targeted therapy options for pancreatic cancer to improve survival.
Background
Pancreatic cancer has one of the highest mortality rates due to its typical late-stage diagnosis and subsequent limited surgical options. However, recent advances in molecular profiling offer hope for targeted therapies. We present a case of locally advanced pancreatic adenocarcinoma which progressed despite surgery and chemotherapy yet showed a positive respond to Alectinib.
Case Description
A 79-year-old male with medical history of tobacco use and ulcerative colitis presented to the clinic with 15lb unintentional weight loss over the past few months in 04/2021. Computed tomography (CT) showed dilated common bile duct due to 2.2 x 1.9 x 1.7 cm mass with no metastatic disease. Biopsy was consistent with pancreatic adenocarcinoma and patient completed 6 cycles of dose-reduced neoadjuvant gemcitabine and paclitaxel in late 2021 due to his severe neuropathy and ECOG. Subsequent CT and PET-CT showed stable disease prior to undergoing pylorus-sparing pancreatoduodenectomy and cholecystectomy with portal vein resection in 05/2022 with surgical pathology grading yPT4N2cM0. The follow- up PET scan in 09/2022 revealed new pulmonary and liver metastases, along with increased uptake in the pancreatic region, suggesting recurrent disease. Next generation sequencing (NGS) identified an ELM4-ALK chromosomal rearrangement on the surgical pathology. Given the patient’s cancer progression and concerns about chemotherapy tolerance, Alectinib, a second-generation ALK inhibitor more commonly used in lung cancer, was considered as a treatment option. Patient began Alectinib 10/2022 with no significant side effects and PET scan on 03/2023 and 06/2023 showing resolution of his lung nodules and liver lesions. Patient remained on Alectinib until he transitioned to hospice after an ischemic stroke in 03/2024.
Discussion
Pancreatic cancer urgently requires novel therapies as about 25% of patients harbor actionable molecular alterations that have led to the success of targeted therapies. ALK fusion genes are identified in multiple cancers, but the prevalence is only 0.16% in pancreatic ductal adenocarcinoma. Alectinib provided an extended progression free survival compared with standard chemotherapy in our patient. ALK inhibitors may demonstrate a remarkable response in metastatic pancreatic cancer even in poor candidates for standard chemotherapy highlighting the emphasis of NGS and targeted therapy options for pancreatic cancer to improve survival.
Background
Pancreatic cancer has one of the highest mortality rates due to its typical late-stage diagnosis and subsequent limited surgical options. However, recent advances in molecular profiling offer hope for targeted therapies. We present a case of locally advanced pancreatic adenocarcinoma which progressed despite surgery and chemotherapy yet showed a positive respond to Alectinib.
Case Description
A 79-year-old male with medical history of tobacco use and ulcerative colitis presented to the clinic with 15lb unintentional weight loss over the past few months in 04/2021. Computed tomography (CT) showed dilated common bile duct due to 2.2 x 1.9 x 1.7 cm mass with no metastatic disease. Biopsy was consistent with pancreatic adenocarcinoma and patient completed 6 cycles of dose-reduced neoadjuvant gemcitabine and paclitaxel in late 2021 due to his severe neuropathy and ECOG. Subsequent CT and PET-CT showed stable disease prior to undergoing pylorus-sparing pancreatoduodenectomy and cholecystectomy with portal vein resection in 05/2022 with surgical pathology grading yPT4N2cM0. The follow- up PET scan in 09/2022 revealed new pulmonary and liver metastases, along with increased uptake in the pancreatic region, suggesting recurrent disease. Next generation sequencing (NGS) identified an ELM4-ALK chromosomal rearrangement on the surgical pathology. Given the patient’s cancer progression and concerns about chemotherapy tolerance, Alectinib, a second-generation ALK inhibitor more commonly used in lung cancer, was considered as a treatment option. Patient began Alectinib 10/2022 with no significant side effects and PET scan on 03/2023 and 06/2023 showing resolution of his lung nodules and liver lesions. Patient remained on Alectinib until he transitioned to hospice after an ischemic stroke in 03/2024.
Discussion
Pancreatic cancer urgently requires novel therapies as about 25% of patients harbor actionable molecular alterations that have led to the success of targeted therapies. ALK fusion genes are identified in multiple cancers, but the prevalence is only 0.16% in pancreatic ductal adenocarcinoma. Alectinib provided an extended progression free survival compared with standard chemotherapy in our patient. ALK inhibitors may demonstrate a remarkable response in metastatic pancreatic cancer even in poor candidates for standard chemotherapy highlighting the emphasis of NGS and targeted therapy options for pancreatic cancer to improve survival.
VA Ann Arbor Immunotherapy Stewardship Program
Purpose
To compare vial utilization and spending between fixed and weight-based dosing of pembrolizumab in Veterans. Promote and assess pembrolizumab extended interval dosing.
Background
FDA approved pembrolizumab label change from weight-based to fixed dosing without evidence of fixed-dosing’s superiority. Retrospective studies demonstrate equivalent outcomes for 2 mg/kg every 3 weeks (Q3W), 200 mg Q3W, 4 mg/kg every 6 weeks (Q6W), and 400 mg Q6W.
Methods
In July 2024 VAAAHS (VA Ann Arbor Healthcare System) initiated an immunotherapy stewardship quality improvement program to deprescribe unnecessary pembrolizumab units and promote extended-interval dosing. Specific interventions included order template modification and targeted outreach to key stakeholders.
Data Analysis
All pembrolizumab doses administered at VAAAHS between July 1, 2024 (launch) and March 31, 2025 (data cutoff) were extracted from EHR. Drug utilization, spending, and healthcare contact hours averted were compared to a fixed-dosing counterfactual.
Results
Sixty-three Veterans received 286 total pembrolizumab doses, of which 107 (37.4%) were Q6W and 179 (62.6%) were Q3W. In total, 741 vials were utilized, against expectation of 786 (5.7% reduction), reflecting approximately $182,000 in savings (annualized, $243,000) and 86.5% of the theoretical maximum savings were captured. Q6W’s share of all doses rose from 27.3% in July 2024 to 53.8% in March 2025. Amongst monotherapy, Q6W’s share rose from 60.0% in July 2024 to 86.7% in March 2025. Q6W adoption saved 381 Veteran-healthcare contact hours, not including travel time.
Conclusions
Stewardship efforts reduced unnecessary pembrolizumab utilization and spending while saving Veterans and VAAAHS providers’ time. Continued provider reinforcement, preparation for Oracle/ Cerner implementation, VISN expansion, refinement of pembrolizumab dose-banding, and development of dose bands for other immunotherapies are underway.
Significance
National implementation would improve Veteran convenience and quality of life, enable reductions in drug and resource costs, and enhance clinic throughput.
Purpose
To compare vial utilization and spending between fixed and weight-based dosing of pembrolizumab in Veterans. Promote and assess pembrolizumab extended interval dosing.
Background
FDA approved pembrolizumab label change from weight-based to fixed dosing without evidence of fixed-dosing’s superiority. Retrospective studies demonstrate equivalent outcomes for 2 mg/kg every 3 weeks (Q3W), 200 mg Q3W, 4 mg/kg every 6 weeks (Q6W), and 400 mg Q6W.
Methods
In July 2024 VAAAHS (VA Ann Arbor Healthcare System) initiated an immunotherapy stewardship quality improvement program to deprescribe unnecessary pembrolizumab units and promote extended-interval dosing. Specific interventions included order template modification and targeted outreach to key stakeholders.
Data Analysis
All pembrolizumab doses administered at VAAAHS between July 1, 2024 (launch) and March 31, 2025 (data cutoff) were extracted from EHR. Drug utilization, spending, and healthcare contact hours averted were compared to a fixed-dosing counterfactual.
Results
Sixty-three Veterans received 286 total pembrolizumab doses, of which 107 (37.4%) were Q6W and 179 (62.6%) were Q3W. In total, 741 vials were utilized, against expectation of 786 (5.7% reduction), reflecting approximately $182,000 in savings (annualized, $243,000) and 86.5% of the theoretical maximum savings were captured. Q6W’s share of all doses rose from 27.3% in July 2024 to 53.8% in March 2025. Amongst monotherapy, Q6W’s share rose from 60.0% in July 2024 to 86.7% in March 2025. Q6W adoption saved 381 Veteran-healthcare contact hours, not including travel time.
Conclusions
Stewardship efforts reduced unnecessary pembrolizumab utilization and spending while saving Veterans and VAAAHS providers’ time. Continued provider reinforcement, preparation for Oracle/ Cerner implementation, VISN expansion, refinement of pembrolizumab dose-banding, and development of dose bands for other immunotherapies are underway.
Significance
National implementation would improve Veteran convenience and quality of life, enable reductions in drug and resource costs, and enhance clinic throughput.
Purpose
To compare vial utilization and spending between fixed and weight-based dosing of pembrolizumab in Veterans. Promote and assess pembrolizumab extended interval dosing.
Background
FDA approved pembrolizumab label change from weight-based to fixed dosing without evidence of fixed-dosing’s superiority. Retrospective studies demonstrate equivalent outcomes for 2 mg/kg every 3 weeks (Q3W), 200 mg Q3W, 4 mg/kg every 6 weeks (Q6W), and 400 mg Q6W.
Methods
In July 2024 VAAAHS (VA Ann Arbor Healthcare System) initiated an immunotherapy stewardship quality improvement program to deprescribe unnecessary pembrolizumab units and promote extended-interval dosing. Specific interventions included order template modification and targeted outreach to key stakeholders.
Data Analysis
All pembrolizumab doses administered at VAAAHS between July 1, 2024 (launch) and March 31, 2025 (data cutoff) were extracted from EHR. Drug utilization, spending, and healthcare contact hours averted were compared to a fixed-dosing counterfactual.
Results
Sixty-three Veterans received 286 total pembrolizumab doses, of which 107 (37.4%) were Q6W and 179 (62.6%) were Q3W. In total, 741 vials were utilized, against expectation of 786 (5.7% reduction), reflecting approximately $182,000 in savings (annualized, $243,000) and 86.5% of the theoretical maximum savings were captured. Q6W’s share of all doses rose from 27.3% in July 2024 to 53.8% in March 2025. Amongst monotherapy, Q6W’s share rose from 60.0% in July 2024 to 86.7% in March 2025. Q6W adoption saved 381 Veteran-healthcare contact hours, not including travel time.
Conclusions
Stewardship efforts reduced unnecessary pembrolizumab utilization and spending while saving Veterans and VAAAHS providers’ time. Continued provider reinforcement, preparation for Oracle/ Cerner implementation, VISN expansion, refinement of pembrolizumab dose-banding, and development of dose bands for other immunotherapies are underway.
Significance
National implementation would improve Veteran convenience and quality of life, enable reductions in drug and resource costs, and enhance clinic throughput.
Assessing the Impact of Antidepressants on Cancer Treatment: A Retrospective Analysis of 14 Antineoplastic Agents
Assessing the Impact of Antidepressants on Cancer Treatment: A Retrospective Analysis of 14 Antineoplastic Agents
Cancer patients experience depression at rates > 5 times that of the general population.1-11 Despite an increase in palliative care use, depression rates continued to rise.2-4 Between 5% to 16% of outpatients, 4% to 14% of inpatients, and up to 49% of patients receiving palliative care experience depression.5 This issue also impacts families and caregivers.1 A 2021 meta-analysis found that 23% of active military personnel and 20% of veterans experience depression.11
Antidepressants approved by the US Food and Drug Administration (FDA) target the serotonin, norepinephrine, or dopamine systems and include boxed warnings about an increased risk of suicidal thoughts in adults aged 18 to 24 years.12,13 These medications are categorized into several classes: monoamine oxidase inhibitors (MAOIs), tricyclic antidepressants (TCAs), tetracyclic antidepressants (TeCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), serotonin receptor modulators (SRMs), serotonin-melatonin receptor antagonists (SMRAs), and N—methyl-D-aspartate receptor antagonists (NMDARAs).14,15 The first FDA-approved antidepressants, iproniazid (an MAOI) and imipramine (a TCA) laid the foundation for the development of newer classes like SSRIs and SNRIs.15-17
Older antidepressants such as MAOIs and TCAs are used less due to their adverse effects (AEs) and drug interactions. MAOIs, such as iproniazid, selegiline, moclobemide, tranylcypromine, isocarboxazid, and phenelzine, have numerous AEs and drug interactions, making them unsuitable for first- or second-line treatment of depression.14,18-21 TCAs such as doxepin, amitriptyline, nortriptyline, imipramine, desipramine, clomipramine, trimipramine, protriptyline, maprotiline, and amoxapine have a narrow therapeutic index requiring careful monitoring for signs of toxicity such as QRS widening, tremors, or confusion. Despite the issues, TCAs are generally classified as second-line agents for major depressive disorder (MDD). TCAs have off-label uses for migraine prophylaxis, treatment of obsessive-compulsive disorder (OCD), insomnia, and chronic pain management first-line.14,22-29
Newer antidepressants, including TeCAs and NDRIs, are typically more effective, but also come with safety concerns. TeCAs like mirtazapine interact with several medications, including MAOIs, serotonin-increasing drugs, alcohol, cannabidiol, and marijuana. Mirtazapine is FDA-approved for the treatment of moderate to severe depression in adults. It is also used off-label to treat insomnia, panic disorder, posttraumatic stress disorder (PTSD), generalized anxiety disorder (GAD), social anxiety disorder (SAD), headaches, and migraines. Compared to other antidepressants, mirtazapine is effective for all stages of depression and addresses a broad range of related symptoms.14,30-34 NDRIs, such as bupropion, also interact with various medications, including MAOIs, other antidepressants, stimulants, and alcohol. Bupropion is FDA-approved for smoking cessation and to treat depression and SAD. It is also used off-label for depression- related bipolar disorder or sexual dysfunction, attention-deficit/hyperactivity disorder (ADHD), and obesity.14,35-42
SSRIs, SNRIs, and SRMs should be used with caution. SSRIs such as sertraline, citalopram, escitalopram, fluoxetine, paroxetine, and fluvoxamine are first-line treatments for depression and various psychiatric disorders due to their safety and efficacy. Common AEs of SSRIs include sexual dysfunction, sleep disturbances, weight changes, and gastrointestinal (GI) issues. SSRIs can prolong the QT interval, posing a risk of life-threatening arrhythmia, and may interact with other medications, necessitating treatment adjustments. The FDA approved SSRIs for MDD, GAD, bulimia nervosa, bipolar depression, OCD, panic disorder, premenstrual dysphoric disorder, treatment-resistant depression, PTSD, and SAD. Off-label uses include binge eating disorder, body dysmorphic disorder, fibromyalgia, premature ejaculation, paraphilias, autism, Raynaud phenomenon, and vasomotor symptoms associated with menopause. Among SSRIs, sertraline and escitalopram are noted for their effectiveness and tolerability.14,43-53
SNRIs, including duloxetine, venlafaxine, desvenlafaxine, milnacipran, and levomilnacipran, may increase bleeding risk, especially when taken with blood thinners. They can also elevate blood pressure, which may worsen if combined with stimulants. SNRIs may interact with other medications that affect serotonin levels, increasing the risk of serotonin syndrome when taken with triptans, pain medications, or other antidepressants.14 Desvenlafaxine has been approved by the FDA (but not by the European Medicines Agency).54-56 Duloxetine is FDA-approved for the treatment of depression, neuropathic pain, anxiety disorders, fibromyalgia, and musculoskeletal disorders. It is used off-label to treat chemotherapy-induced peripheral neuropathy and stress urinary incontinence.57-61 Venlafaxine is FDA-approved for depression, SAD, and panic disorder, and is prescribed off-label to treat ADHD, neuropathy, fibromyalgia, cataplexy, and PTSD, either alone or in combination with other medications.62,63 Milnacipran is not approved for MDD; levomilnacipran received approval in 2013.64
SRMs such as trazodone, nefazodone, vilazodone, and vortioxetine also function as serotonin reuptake inhibitors.14,15 Trazodone is FDA-approved for MDD. It has been used off-label to treat anxiety, Alzheimer disease, substance misuse, bulimia nervosa, insomnia, fibromyalgia, and PTSD when first-line SSRIs are ineffective. A notable AE of trazodone is orthostatic hypotension, which can lead to dizziness and increase the risk of falls, especially in geriatric patients.65-70 Nefazodone was discontinued in Europe in 2003 due to rare cases of liver toxicity but remains available in the US.71-74 Vilazodone and vortioxetine are FDA-approved.
The latest classes of antidepressants include SMRAs and NMDARAs.14 Agomelatine, an SMRA, was approved in Europe in 2009 but rejected by the FDA in 2011 due to liver toxicity.75 NMDARAs like esketamine and a combination of dextromethorphan and bupropion received FDA approval in 2019 and 2022, respectively.76,77
This retrospective study analyzes noncancer drugs used during systemic chemotherapy based on a dataset of 14 antineoplastic agents. It sought to identify the most dispensed noncancer drug groups, discuss findings, compare patients with and without antidepressant prescriptions, and examine trends in antidepressant use from 2002 to 2023. This analysis expands on prior research.78-81
Methods
The Walter Reed National Military Medical Center Institutional Review Board approved the study protocol and ensured compliance with the Health Insurance Portability and Accountability Act as an exempt protocol. The Joint Pathology Center (JPC) of the US Department of Defense (DoD) Cancer Registry Program and Military Health System (MHS) data experts from the Comprehensive Ambulatory/Professional Encounter Record (CAPER) and Pharmacy Data Transaction Service (PDTS) provided data for the analysis.
Data Sources
The JPC DoD Cancer Registry Program contains data from 1998 to 2024. CAPER and PDTS are part of the MHS Data Repository/Management Analysis and Reporting Tool database. Each observation in CAPER represents an ambulatory encounter at a military treatment facility (MTF). CAPER records are available from 2003 to 2024. PDTS records are available from 2002 to 2004. Each observation in PDTS represents a prescription filled for an MHS beneficiary, excluding those filled at international civilian pharmacies and inpatient pharmacy prescriptions.
This cross-sectional analysis requested data extraction for specific cancer drugs from the DoD Cancer Registry, focusing on treatment details, diagnosis dates, patient demographics, and physicians’ comments on AEs. After identifying patients, CAPER was used to identify additional health conditions. PDTS was used to compile a list of prescription medications filled during systemic cancer treatment or < 2 years postdiagnosis.
The 2016 Surveillance, Epidemiology, and End Results Program Coding and Staging Manual and International Classification of Diseases for Oncology, 3rd edition, 1st revision, were used to decode disease and cancer types.82,83 Data sorting and analysis were performed using Microsoft Excel. The percentage for the total was calculated by using the number of patients or data available within the subgroup divided by the total number of patients or data variables. To compare the mean number of dispensed antidepressants to those without antidepressants, a 2-tailed, 2-sample z test was used to calculate the P value and determine statistical significance (P < .05) using socscistatistics.com.
Data were extracted 3 times between 2021 and 2023. The initial 2021 protocol focused on erlotinib and gefitinib. A modified protocol in 2022 added paclitaxel, cisplatin, docetaxel, pemetrexed, and crizotinib; further modification in 2023 included 8 new antineoplastic agents and 2 anticoagulants. Sotorasib has not been prescribed in the MHS, and JPC lacks records for noncancer drugs. The 2023 dataset comprised 2210 patients with cancer treated with 14 antineoplastic agents; 2104 had documented diagnoses and 2113 had recorded prescriptions. Data for erlotinib, gefitinib, and paclitaxel have been published previously.78,79
Results
Of 2113 patients with recorded prescriptions, 1297 patients (61.4%) received 109 cancer drugs, including 96 antineoplastics, 7 disease-modifying antirheumatic agents, 4 biologic response modifiers, and 2 calcitonin gene-related peptides. Fourteen antineoplastic agents had complete data from JPC, while others were noted for combination therapies or treatment switches from the PDTS (Table 1). Seventy-six cancer drugs were prescribed with antidepressants in 489 patients (eAppendix).

The JPC provided 2242 entries for 2210 patients, ranging in age from 2 months to 88 years (mean, 56 years), documenting treatment from September 1988 to January 2023. Thirty-two patients had duplicate entries due to multiple cancer locations or occurrences. Of the 2242 patients, 1541 (68.7%) were aged > 50 years, 975 patients (43.5%) had cancers that were stage III or IV, and 1267 (56.5%) had cancers that were stage 0, I, II, or not applicable/unknown. There were 51 different types of cancer: breast, lung, testicular, endometrial, and ovarian were most common (n ≥ 100 patients). Forty-two cancer types were documented among 750 patients prescribed antidepressants (Table 2).

The CAPER database recorded 8882 unique diagnoses for 2104 patients, while PDTS noted 1089 unique prescriptions within 273 therapeutic codes for 2113 patients. Nine therapeutic codes (opiate agonists, adrenals, cathartics-laxatives, nonsteroidal anti-inflammatory agents, antihistamines for GI conditions, 5-HT3 receptor antagonists, analgesics and antipyretic miscellanea, antineoplastic agents, and proton-pump inhibitors) and 8 drugs (dexamethasone, prochlorperazine, ondansetron, docusate, acetaminophen, ibuprofen, oxycodone, and polyethylene glycol 3350) were associated with > 1000 patients (≥ 50%). Patients had between 1 and 275 unique health conditions and filled 1 to 108 prescriptions. The mean (SD) number of diagnoses and prescriptions was 50 (28) and 29 (12), respectively. Of the 273 therapeutic codes, 30 groups were analyzed, with others categorized into miscellaneous groups such as lotions, vaccines, and devices. Significant differences in mean number of prescriptions were found for patients taking antidepressants compared to those not (P < .05), except for anticonvulsants and antipsychotics (P = .12 and .09, respectively) (Table 3).

Antidepressants
Of the 2113 patients with recorded prescriptions, 750 (35.5%) were dispensed 17 different antidepressants. Among these 17 antidepressants, 183 (8.7%) patients received duloxetine, 158 (7.5%) received venlafaxine, 118 (5.6%) received trazodone, and 107 (5.1%) received sertraline (Figure 1, Table 4). Of the 750 patients, 509 (67.9%) received 1 antidepressant, 168 (22.4%) received 2, 60 (8.0%) received 3, and 13 (1.7%) received > 3. Combinations varied, but only duloxetine and trazodone were prescribed to > 10 patients.



Antidepressants were prescribed annually at an overall mean (SD) rate of 23% (5%) from 2003 to 2022 (Figure 2). Patients on antidepressants during systemic therapy had a greater number of diagnosed medical conditions and received more prescription medications compared to those not taking antidepressants (P < .001) (Figure 3). The 745 patients taking antidepressants in CAPER data had between 1 and 275 diagnosed medical issues, with a mean (SD) of 55 (31) vs a range of 1 to 209 and a mean (SD) of 46 (26) for the 1359 patients not taking antidepressants. The 750 patients on antidepressants in PDTS data had between 8 and 108 prescriptions dispensed, with a mean (SD) of 32 (12), vs a range of 1 to 65 prescriptions and a mean (SD) of 29 (12) for 1363 patients not taking antidepressants.


Discussion
The JPC DoD Cancer Registry includes information on cancer types, stages, treatment regimens, and physicians’ notes, while noncancer drugs are sourced from the PDTS database. The pharmacy uses a different documentation system, leading to varied classifications.
Database reliance has its drawbacks. For example, megestrol is coded as a cancer drug, although it’s primarily used for endometrial or gynecologic cancers. Many drugs have multiple therapeutic codes assigned to them, including 10 antineoplastic agents: diclofenac, Bacillus Calmette-Guérin (BCG), megestrol acetate, tamoxifen, anastrozole, letrozole, leuprolide, goserelin, degarelix, and fluorouracil. Diclofenac, BCG, and mitomycin have been repurposed for cancer treatment.84-87 From 2003 to 2023, diclofenac was prescribed to 350 patients for mild-to-moderate pain, with only 2 patients receiving it for cancer in 2018. FDA-approved for bladder cancer in 1990, BCG was prescribed for cancer treatment for 1 patient in 2021 after being used for vaccines between 2003 and 2018. Tamoxifen, used for hormone receptor-positive breast cancer from 2004 to 2017 with 53 patients, switched to estrogen agonist-antagonists from 2017 to 2023 with 123 patients. Only a few of the 168 patients were prescribed tamoxifen using both codes.88-91 Anastrozole and letrozole were coded as antiestrogens for 7 and 18 patients, respectively, while leuprolide and goserelin were coded as gonadotropins for 59 and 18 patients. Degarelix was coded as antigonadotropins, fluorouracil as skin and mucous membrane agents miscellaneous, and megestrol acetate as progestins for 7, 6, and 3 patients, respectively. Duloxetine was given to 186 patients, primarily for depression from 2005 to 2023, with 7 patients treated for fibromyalgia from 2022 to 2023.
Antidepressants Observed
Tables 1 and 5 provide insight into the FDA approval of 14 antineoplastics and antidepressants and their CYP metabolic pathways.92-122 In Table 4, the most prescribed antidepressant classes are SNRIs, SRMs, SSRIs, TeCAs, NDRIs, and TCAs. This trend highlights a preference for newer medications with weak CYP inhibition. A total of 349 patients were prescribed SSRIs, 343 SNRIs, 119 SRMs, 109 TCAs, 83 TeCAs, and 79 NDRIs. MAOIs, SMRAs, and NMDARAs were not observed in this dataset. While there are instances of dextromethorphan-bupropion and sertraline-escitalopram being dispensed together, it remains unclear whether these were NMDARA combinations.
Among the 14 specific antineoplastic agents, 10 are metabolized by CYP isoenzymes, primarily CYP3A4. Duloxetine neither inhibits nor is metabolized by CYP3A4, a reason it is often recommended, following venlafaxine.
Both duloxetine and venlafaxine are used off-label for chemotherapy-induced peripheral neuropathy related to paclitaxel and docetaxel. According to the CYP metabolized pathway, duloxetine tends to have more favorable DDIs than venlafaxine. In PDTS data, 371 patients were treated with paclitaxel and 180 with docetaxel, with respective antidepressant prescriptions of 156 and 70. Of the 156 patients dispensed paclitaxel, 62 (40%) were dispensed with duloxetine compared to 43 (28%) with venlafaxine. Of the 70 patients dispensed docetaxel, 23 (33%) received duloxetine vs 24 (34%) with venlafaxine.
Of 85 patients prescribed duloxetine, 75 received it with either paclitaxel or docetaxel (5 received both). Five patients had documented AEs (1 neuropathy related). Of 67 patients prescribed venlafaxine, 66 received it with either paclitaxel or docetaxel. Two patients had documented AEs (1 was neuropathy related, the same patient who received duloxetine). Of the 687 patients treated with paclitaxel and 337 with docetaxel in all databases, 4 experienced neuropathic AEs from both medications.79
Antidepressants can increase the risk of bleeding, especially when combined with blood thinners, and may elevate blood pressure, particularly alongside stimulants. Of the 554 patients prescribed 9 different anticoagulants, enoxaparin, apixaban, and rivaroxaban were the most common (each > 100 patients). Among these, 201 patients (36%) received both anticoagulants and antidepressants: duloxetine for 64 patients, venlafaxine for 30, trazodone for 35, and sertraline for 26. There were no data available to assess bleeding rates related to the evaluation of DDIs between these medication classes.
Antidepressants can be prescribed for erectile dysfunction. Of the 148 patients prescribed an antidepressant for erectile dysfunction, duloxetine, trazodone, and mirtazapine were the most common. Antidepressant preferences varied by cancer type. Duloxetine was the only antidepressant used for all types of cancer. Venlafaxine, duloxetine, trazodone, sertraline, and escitalopram were the most prescribed antidepressants for breast cancer, while duloxetine, mirtazapine, citalopram, sertraline, and trazodone were the most prescribed for lung cancer. Sertraline, duloxetine, trazodone, amitriptyline, and escitalopram were most common for testicular cancer. Duloxetine, venlafaxine, trazodone, amitriptyline, and sertraline were the most prescribed for endometrial cancer, while duloxetine, venlafaxine, amitriptyline, citalopram, and sertraline were most prescribed for ovarian cancer.
The broadness of International Statistical Classification of Diseases, Tenth Revision codes made it challenging to identify nondepression diagnoses in the analyzed population. However, if all antidepressants were prescribed to treat depression, service members with cancer exhibited a higher depression rate (35%) than the general population (25%). Of 2104 patients, 191 (9.1%) had mood disorders, and 706 (33.6%) had mental disorders: 346 (49.0%) had 1 diagnosis, and 360 (51.0%) had multiple diagnoses. The percentage of diagnoses varied yearly, with notable drops in 2003, 2007, 2011, 2014, and 2018, and peaks in 2006, 2008, 2013, 2017, and 2022. This fluctuation was influenced by events like the establishment of PDTS in 2002, the 2008 economic recession, a hospital relocation in 2011, the 2014 Ebola outbreak, and the COVID-19 pandemic. Although the number of patients receiving antidepressants increased from 2019 to 2022, the overall percentage of patients receiving them did not significantly change from 2003 to 2022, aligning with previous research.5,125
Many medications have potential uses beyond what is detailed in the prescribing information. Antidepressants can relieve pain, while pain medications may help with depression. Opioids were once thought to effectively treat depression, but this perspective has changed with a greater understanding of their risks, including misuse.126-131 Pain is a severe and often unbearable AE of cancer. Of 2113 patients, 92% received opioids; 34% received both opioids and antidepressants; 2% received only antidepressants; and 7% received neither. This study didn’t clarify whether those on opioids alone recognized their depression or if those on both were aware of their dependence. While SSRIs are generally not addictive, they can lead to physical dependence, and any medication can be abused if not managed properly.132-134
Conclusions
This retrospective study analyzes data from antineoplastic agents used in systemic cancer treatment between 1988 and 2023, with a particular focus on the use of antidepressants. Data on antidepressant prescriptions are incomplete and specific to these agents, which means the findings cannot be generalized to all antidepressants. Hence, the results indicate that patients taking antidepressants had more diagnosed health issues and received more medications compared to patients who were not on these drugs.
This study underscores the need for further research into the effects of antidepressants on cancer treatment, utilizing all data from the DoD Cancer Registry. Future research should explore DDIs between antidepressants and other cancer and noncancer medications, as this study did not assess AE documentation, unlike in studies involving erlotinib, gefitinib, and paclitaxel.78,79 Further investigation is needed to evaluate the impact of discontinuing antidepressant use during cancer treatment. This comprehensive overview provides insights for clinicians to help them make informed decisions regarding the prescription of antidepressants in the context of cancer treatment.
- National Cancer Institute. Depression (PDQ)-Health Professional Version. Updated July 25, 2024. Accessed April 4, 2025. https://www.cancer.gov/about-cancer/coping/feelings/depression-hp-pdq
- Krebber AM, Buffart LM, Kleijn G, et al. Prevalence of depression in cancer patients: a meta-analysis of diagnostic interviews and self-report instruments. Psychooncology. 2014;23(2):121-130. doi:10.1002/pon.3409
- Xiang X, An R, Gehlert S. Trends in antidepressant use among U.S. cancer survivors, 1999-2012. Psychiatr Serv. 2015;66(6):564. doi:10.1176/appi.ps.201500007
- Hartung TJ, Brähler E, Faller H, et al. The risk of being depressed is significantly higher in cancer patients than in the general population: Prevalence and severity of depressive symptoms across major cancer types. Eur J Cancer. 2017;72:46-53. doi:10.1016/j.ejca.2016.11.017
- Walker J, Holm Hansen C, Martin P, et al. Prevalence of depression in adults with cancer: a systematic review. Ann Oncol. 2013;24(4):895-900. doi:10.1093/annonc/mds575
- Pitman A, Suleman S, Hyde N, Hodgkiss A. Depression and anxiety in patients with cancer. BMJ. 2018;361:k1415. doi:10.1136/bmj.k1415
- Kisely S, Alotiby MKN, Protani MM, Soole R, Arnautovska U, Siskind D. Breast cancer treatment disparities in patients with severe mental illness: a systematic review and meta-analysis. Psychooncology. 2023;32(5):651-662. doi:10.1002/pon.6120
- Massie MJ. Prevalence of depression in patients with cancer. J Natl Cancer Inst Monogr. 2004;(32):57-71. doi:10.1093/jncimonographs/lgh014
- Rodin G, Katz M, Lloyd N, Green E, Mackay JA, Wong RK. Treatment of depression in cancer patients. Curr Oncol. 2007;14(5):180-188. doi:10.3747/co.2007.146
- Wilson KG, Chochinov HM, Skirko MG, et al. Depression and anxiety disorders in palliative cancer care. J Pain Symptom Manage. 2007;33(2):118-129. doi:10.1016/j.jpainsymman.2006.07.016
- Moradi Y, Dowran B, Sepandi M. The global prevalence of depression, suicide ideation, and attempts in the military forces: a systematic review and meta-analysis of cross sectional studies. BMC Psychiatry. 2021;21(1):510. doi:10.1186/s12888-021-03526-2
- Lu CY, Zhang F, Lakoma MD, et al. Changes in antidepressant use by young people and suicidal behavior after FDA warnings and media coverage: quasi-experimental study. BMJ. 2014;348:g3596. doi:10.1136/bmj.g3596
- Friedman RA. Antidepressants’ black-box warning--10 years later. N Engl J Med. 2014;371(18):1666-1668. doi:10.1056/NEJMp1408480
- Sheffler ZM, Patel P, Abdijadid S. Antidepressants. In: StatPearls. StatPearls Publishing. Updated May 26, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK538182/
- Miller JJ. Antidepressants, Part 1: 100 Years and Counting. Accessed April 4, 2025. Psychiatric Times. https:// www.psychiatrictimes.com/view/antidepressants-part-1-100-years-and-counting
- Hillhouse TM, Porter JH. A brief history of the development of antidepressant drugs: from monoamines to glutamate. Exp Clin Psychopharmacol. 2015;23(1):1-21. doi:10.1037/a0038550
- Mitchell PB, Mitchell MS. The management of depression. Part 2. The place of the new antidepressants. Aust Fam Physician. 1994;23(9):1771-1781.
- Sabri MA, Saber-Ayad MM. MAO Inhibitors. In: Stat- Pearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557395/
- Sub Laban T, Saadabadi A. Monoamine Oxidase Inhibitors (MAOI). In: StatPearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK539848/
- Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract. 2004;10(4):239-248. doi:10.1097/00131746-200407000-00005
- Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03
- Moraczewski J, Awosika AO, Aedma KK. Tricyclic Antidepressants. In: StatPearls. StatPearls Publishing. Updated August 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557791/
- Almasi A, Patel P, Meza CE. Doxepin. In: StatPearls. StatPearls Publishing. Updated February 14, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK542306/
- Thour A, Marwaha R. Amitriptyline. In: StatPearls. Stat- Pearls Publishing. Updated July 18, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK537225/
- Radley DC, Finkelstein SN, Stafford RS. Off-label prescribing among office-based physicians. Arch Intern Med. 2006;166(9):1021-1026. doi:10.1001/archinte.166.9.1021
- Tesfaye S, Sloan G, Petrie J, et al. Comparison of amitriptyline supplemented with pregabalin, pregabalin supplemented with amitriptyline, and duloxetine supplemented with pregabalin for the treatment of diabetic peripheral neuropathic pain (OPTION-DM): a multicentre, double-blind, randomised crossover trial. Lancet. 2022;400(10353):680- 690. doi:10.1016/S0140-6736(22)01472-6
- Farag HM, Yunusa I, Goswami H, Sultan I, Doucette JA, Eguale T. Comparison of amitriptyline and US Food and Drug Administration-approved treatments for fibromyalgia: a systematic review and network metaanalysis. JAMA Netw Open. 2022;5(5):e2212939. doi:10.1001/jamanetworkopen.2022.12939
- Merwar G, Gibbons JR, Hosseini SA, Saadabadi A. Nortriptyline. In: StatPearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482214/
- Fayez R, Gupta V. Imipramine. In: StatPearls. StatPearls Publishing. Updated May 22, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557656/
- Jilani TN, Gibbons JR, Faizy RM, Saadabadi A. Mirtazapine. In: StatPearls. StatPearls Publishing. Updated August 28, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK519059/
- Nutt DJ. Tolerability and safety aspects of mirtazapine. Hum Psychopharmacol. 2002;17 Suppl 1:S37-S41. doi:10.1002/hup.388
- Gandotra K, Chen P, Jaskiw GE, Konicki PE, Strohl KP. Effective treatment of insomnia with mirtazapine attenuates concomitant suicidal ideation. J Clin Sleep Med. 2018;14(5):901-902. doi:10.5664/jcsm.7142
- Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249-264. doi:10.1111/j.1527-3458.2001.tb00198.x
- Wang SM, Han C, Bahk WM, et al. Addressing the side effects of contemporary antidepressant drugs: a comprehensive review. Chonnam Med J. 2018;54(2):101-112. doi:10.4068/cmj.2018.54.2.101
- Huecker MR, Smiley A, Saadabadi A. Bupropion. In: StatPearls. StatPearls Publishing. Updated April 9, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470212/
- Hsieh MT, Tseng PT, Wu YC, et al. Effects of different pharmacologic smoking cessation treatments on body weight changes and success rates in patients with nicotine dependence: a network meta-analysis. Obes Rev. 2019;20(6):895- 905. doi:10.1111/obr.12835
- Hankosky ER, Bush HM, Dwoskin LP, et al. Retrospective analysis of health claims to evaluate pharmacotherapies with potential for repurposing: association of bupropion and stimulant use disorder remission. AMIA Annu Symp Proc. 2018;2018:1292-1299.
- Livingstone-Banks J, Norris E, Hartmann-Boyce J, West R, Jarvis M, Hajek P. Relapse prevention interventions for smoking cessation. Cochrane Database Syst Rev. 2019; 2(2):CD003999. doi:10.1002/14651858.CD003999.pub5
- Clayton AH, Kingsberg SA, Goldstein I. Evaluation and management of hypoactive sexual desire disorder. Sex Med. 2018;6(2):59-74. doi:10.1016/j.esxm.2018.01.004
- Verbeeck W, Bekkering GE, Van den Noortgate W, Kramers C. Bupropion for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev. 2017;10(10):CD009504. doi:10.1002/14651858.CD009504.pub2
- Ng QX. A systematic review of the use of bupropion for attention-deficit/hyperactivity disorder in children and adolescents. J Child Adolesc Psychopharmacol. 2017;27(2):112-116. doi:10.1089/cap.2016.0124
- Fava M, Rush AJ, Thase ME, et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim Care Companion J Clin Psychiatry. 2005;7(3):106-113. doi:10.4088/pcc.v07n0305
- Chu A, Wadhwa R. Selective Serotonin Reuptake Inhibitors. In: StatPearls. StatPearls Publishing. Updated May 1, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK554406/
- Singh HK, Saadabadi A. Sertraline. In: StatPearls. Stat- Pearls Publishing. Updated February 13, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK547689/
- MacQueen G, Born L, Steiner M. The selective serotonin reuptake inhibitor sertraline: its profile and use in psychiatric disorders. CNS Drug Rev. 2001;7(1):1-24. doi:10.1111/j.1527-3458.2001.tb00188.x
- Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatments meta-analysis. Lancet. 2009;373(9665):746-758. doi:10.1016/S0140-6736(09)60046-5
- Nelson JC. The STAR*D study: a four-course meal that leaves us wanting more. Am J Psychiatry. 2006;163(11):1864-1866. doi:10.1176/ajp.2006.163.11.1864
- Sharbaf Shoar N, Fariba KA, Padhy RK. Citalopram. In: StatPearls. StatPearls Publishing. Updated November 7, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482222/
- Landy K, Rosani A, Estevez R. Escitalopram. In: Stat- Pearls. StatPearls Publishing. Updated November 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557734/
- Cavanah LR, Ray P, Goldhirsh JL, Huey LY, Piper BJ. Rise of escitalopram and the fall of citalopram. medRxiv. Preprint published online May 8, 2023. doi:10.1101/2023.05.07.23289632
- Sohel AJ, Shutter MC, Patel P, Molla M. Fluoxetine. In: StatPearls. StatPearls Publishing. Updated July 4, 2022. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK459223/
- Wong DT, Perry KW, Bymaster FP. Case history: the discovery of fluoxetine hydrochloride (Prozac). Nat Rev Drug Discov. 2005;4(9):764-774. doi:10.1038/nrd1821
- Shrestha P, Fariba KA, Abdijadid S. Paroxetine. In: Stat- Pearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK526022/
- Naseeruddin R, Rosani A, Marwaha R. Desvenlafaxine. In: StatPearls. StatPearls Publishing. Updated July 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK534829
- Lieberman DZ, Massey SH. Desvenlafaxine in major depressive disorder: an evidence-based review of its place in therapy. Core Evid. 2010;4:67-82. doi:10.2147/ce.s5998
- Withdrawal assessment report for Ellefore (desvenlafaxine). European Medicine Agency. 2009. Accessed April 4, 2025. https://www.ema.europa.eu/en/documents/withdrawal-report/withdrawal-assessment-report-ellefore_en.pdf
- Dhaliwal JS, Spurling BC, Molla M. Duloxetine. In: Stat- Pearls. Statpearls Publishing. Updated May 29, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK549806/
- Hershman DL, Lacchetti C, Dworkin RH, et al. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2014;32(18):1941-1967. doi:10.1200/JCO.2013.54.0914
- Sommer C, Häuser W, Alten R, et al. Medikamentöse Therapie des Fibromyalgiesyndroms. Systematische Übersicht und Metaanalyse [Drug therapy of fibromyalgia syndrome. Systematic review, meta-analysis and guideline]. Schmerz. 2012;26(3):297-310. doi:10.1007/s00482-012-1172-2
- Bril V, England J, Franklin GM, et al. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology. 2011;76(20):1758-1765. doi:10.1212/WNL.0b013e3182166ebe
- Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2010;17(9):1113-e88. doi:10.1111/j.1468-1331.2010.02999.x
- Singh D, Saadabadi A. Venlafaxine. In: StatPearls. StatePearls Publishing. Updated February 26, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK535363/
- Sertraline and venlafaxine: new indication. Prevention of recurrent depression: no advance. Prescrire Int. 2005;14(75):19-20.
- Bruno A, Morabito P, S p i n a E , M u s c a t ello MRl. The role of levomilnacipran in the management of major depressive disorder: a comprehensive review. Curr Neuro pharmacol. 2016;14(2):191-199. doi:10.2174/1570159x14666151117122458
- Shin JJ, Saadabadi A. Trazodone. In: StatPearls. StatPearls Publishing. Updated February 29, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470560/
- Khouzam HR. A review of trazodone use in psychiatric and medical conditions. Postgrad Med. 2017;129(1):140-148. doi:10.1080/00325481.2017.1249265
- Smales ET, Edwards BA, Deyoung PN, et al. Trazodone effects on obstructive sleep apnea and non-REM arousal threshold. Ann Am Thorac Soc. 2015;12(5):758-764. doi:10.1513/AnnalsATS.201408-399OC
- Eckert DJ, Malhotra A, Wellman A, White DP. Trazodone increases the respiratory arousal threshold in patients with obstructive sleep apnea and a low arousal threshold. Sleep. 2014;37(4):811-819. doi:10.5665/sleep.3596
- Schatzberg AF, Nemeroff CB, eds. The American Psychiat ric Association Publishing Textbook of Psychopharmacology. 4th ed. American Psychiatric Publishing; 2009.
- Ruxton K, Woodman RJ, Mangoni AA. Drugs with anticholinergic effects and cognitive impairment, falls and allcause mortality in older adults: a systematic review and meta-analysis. Br J Clin Pharmacol. 2015;80(2):209-220. doi:10.1111/bcp.12617
- Nefazodone. In: LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. National Institute of Diabetes and Digestive and Kidney Diseases. Updated March 6, 2020. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK548179/
- Drugs of Current Interest: Nefazodone. WHO Pharmaceuticals Newsletter:(1). 2003(1):7. https://web.archive.org/web/20150403165029/http:/apps.who.int/medicinedocs/en/d/Js4944e/3.html
- Choi S. Nefazodone (serzone) withdrawn because of hepatotoxicity. CMAJ. 2003;169(11):1187.
- Teva Nefazodone Statement. News release. Teva USA. December 20, 2021. Accessed April 4, 2025. https:// www.tevausa.com/news-and-media/press-releases/teva-nefazodone-statement/
- Levitan MN, Papelbaum M, Nardi AE. Profile of agomelatine and its potential in the treatment of generalized anxiety disorder. Neuropsychiatr Dis Treat. 2015;11:1149- 1155. doi:10.2147/NDT.S67470
- Fu DJ, Ionescu DF, Li X, et al. Esketamine nasal spray for rapid reduction of major depressive disorder symptoms in patients who have active suicidal ideation with intent: double-blind, randomized study (ASPIRE I). J Clin Psychiatry. 2020;81(3):19m13191. doi:10.4088/JCP.19m13191
- Iosifescu DV, Jones A, O’Gorman C, et al. Efficacy and safety of AXS-05 (dextromethorphan-bupropion) in patients with major depressive disorder: a phase 3 randomized clinical trial (GEMINI). J Clin Psychiatry. 2022;83(4): 21m14345. doi:10.4088/JCP.21m14345
- Luong TT, Powers CN, Reinhardt BJ, et al. Retrospective evaluation of drug-drug interactions with erlotinib and gefitinib use in the Military Health System. Fed Pract. 2023;40(Suppl 3):S24-S34. doi:10.12788/fp.0401
- Luong TT, Shou KJ, Reinhard BJ, Kigelman OF, Greenfield KM. Paclitaxel drug-drug interactions in the Military Health System. Fed Pract. 2024;41(Suppl 3):S70-S82. doi:10.12788/fp.0499
- Luong TT, Powers CN, Reinhardt BJ, Weina PJ. Preclinical drug-drug interactions (DDIs) of gefitinib with/without losartan and selective serotonin reuptake inhibitors (SSRIs): citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, and venlafaxine. Curr Res Pharmacol Drug Discov. 2022;3:100112. doi:10.1016/j.crphar.2022.100112
- Luong TT, McAnulty MJ, Evers DL, Reinhardt BJ, Weina PJ. Pre-clinical drug-drug interaction (DDI) of gefitinib or erlotinib with cytochrome P450 (CYP) inhibiting drugs, fluoxetine and/or losartan. Curr Res Toxicol. 2021;2:217- 224. doi:10.1016/j.crtox.2021.05.006,
- Adamo M, Dickie L, Ruhl J. SEER program coding and staging manual 2016. National Cancer Institute; 2016. Accessed April 4, 2025. https://seer.cancer.gov/archive/manuals/2016/SPCSM_2016_maindoc.pdf
- World Health Organization. International classification of diseases for oncology (ICD-O). 3rd ed, 1st revision. World Health Organization; 2013. Accessed April 4, 2025. https://apps.who.int/iris/handle/10665/96612
- Simon S. FDA approves Jelmyto (mitomycin gel) for urothelial cancer. American Cancer Society. April 20, 2020. Accessed April 4, 2025. https://www.cancer.org/cancer/latest-news/fda-approves-jelmyto-mitomycin-gel-for-urothelial-cancer.html
- Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing drugs in oncology (ReDO)- diclofenac as an anti-cancer agent. Ecancermedicalscience. 2016;10:610. doi:10.3332/ecancer.2016.610
- Choi S, Kim S, Park J, Lee SE, Kim C, Kang D. Diclofenac: a nonsteroidal anti-inflammatory drug inducing cancer cell death by inhibiting microtubule polymerization and autophagy flux. Antioxidants (Basel). 2022;11(5):1009. doi:10.3390/antiox11051009
- Tontonoz M. The ABCs of BCG: oldest approved immunotherapy gets new explanation. Memorial Sloan Kettering Cancer Center. July 17, 2020. Accessed April 4, 2025. https://www.mskcc.org/news/oldest-approved-immunotherapy-gets-new-explanation
- Jordan VC. Tamoxifen as the first targeted long-term adjuvant therapy for breast cancer. Endocr Relat Cancer. 2014;21(3):R235-R246. doi:10.1530/ERC-14-0092
- Cole MP, Jones CT, Todd ID. A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474. Br J Cancer. 1971;25(2):270-275. doi:10.1038/bjc.1971.33
- Jordan VC. Tamoxifen: a most unlikely pioneering medicine. Nat Rev Drug Discov. 2003;2(3):205-213. doi:10.1038/nrd1031
- Maximov PY, McDaniel RE, Jordan VC. Tamoxifen: Pioneering Medicine in Breast Cancer. Springer Basel; 2013. Accessed April 4, 2025. https://link.springer.com/book/10.1007/978-3-0348-0664-0
- Taxol (paclitaxel). Prescribing information. Bristol-Myers Squibb Company; 2011. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020262s049lbl.pdf
- Abraxane (paclitaxel). Prescribing information. Celgene Corporation; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021660s047lbl.pdf
- Tarceva (erlotinib). Prescribing information. OSI Pharmaceuticals, LLC; 2016. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021743s025lbl.pdf
- Docetaxel. Prescribing information. Sichuan Hyiyu Pharmaceutical Co.; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215813s000lbl.pdf
- Alimta (pemetrexed). Prescribing information. Teva Pharmaceuticals; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/208419s004lbl.pdf
- Tagrisso (osimertinib). Prescribing information. Astra- Zeneca Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208065s021lbl.pdf
- Iressa (gefitinib). Prescribing information. AstraZeneca Pharmaceuticals; 2018. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/206995s003lbl.pdf
- Kadcyla (ado-trastuzumab emtansine). Prescribing information. Genentech, Inc.; 2013. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125427lbl.pdf
- Alecensa (alectinib). Prescribing information. Genetech, Inc.; 2017. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2017/208434s003lbl.pdf
- Xalkori (crizotinib). Prescribing information. Pfizer Laboratories; 2022. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2022/202570s033lbl.pdf
- Lorbrena (lorlatinib). Prescribing information. Pfizer Laboratories; 2018. Accessed April 14, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210868s000lbl.pdf
- Alunbrig (brigatinib). Prescribing information. Takeda Pharmaceutical Company; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208772s008lbl.pdf
- Rozlytrek (entrectinib). Prescribing information. Genentech, Inc.; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212725s000lbl.pdf
- Herceptin (trastuzumab). Prescribing information. Genentech, Inc.; 2010. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/103792s5250lbl.pdf
- Cybalta (duloxetine). Prescribing information. Eli Lilly and Company; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021427s049lbl.pdf
- Effexor XR (venlafaxine). Prescribing information. Pfizer Wyeth Pharmaceuticals Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020699s112lbl.pdf
- Desyrel (trazodone hydrochloride). Prescribing information. Pragma Pharmaceuticals; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/018207s032lbl.pdf
- Sertraline hydrochloride. Prescribing information. Almatica Pharma LLC; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/215133s000lbl.pdf
- Remeron (mirtazapine). Prescribing information. Merck & Co. Inc; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/020415s029,%20021208s019lbl.pdf
- Celexa (citalopram). Prescribing information. Allergan USA Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020822s041lbl.pdf
- information. GlaxoSmithKline; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020358s066lbl.pdf
- Amitriptyline hydrochloride tablet. Prescribing information. Quality Care Products LLC; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0f12f50f-7087-46e7-a2e6-356b4c566c9f/0f12f50f-7087-46e7-a2e6-356b4c566c9f.xml
- Lexapro (escitalopram). Prescribing information. AbbVie Inc; 2023. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/021323s055,021365s039lbl.pdf
- Fluoxetine. Prescribing information. Edgemont Pharmaceutical, LLC; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/202133s004s005lbl.pdf
- Paxil (paroxetine). Prescribing Information. Apotex Inc; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/020031s077lbl.pdf
- Pamelor (nortriptyline HCl). Prescribing information. Mallinckrodt, Inc; 2012. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2012/018012s029,018013s061lbl.pdf
- Silenor (doxepin). Prescribing information. Currax Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022036s006lbl.pdf
- Tofranil-PM (imipramine pamote). Prescribing information. Mallinckrodt, Inc; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/017090s078lbl.pdf
- Norpramin (desipramine hydrochloride). Prescribing information. Sanofi-aventis U.S. LLC; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/014399s069lbl.pdf
- Khedezla (desvenlafaxine). Prescribing information. Osmotical Pharmaceutical US LLC; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/204683s006lbl.pdf
- Nefazodone hydrochloride. Prescribing information. Bryant Ranch Prepack; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0bd4c34a-4f43-4c84-8b98-1d074cba97d5/0bd4c34a-4f43-4c84-8b98-1d074cba97d5.xml
- Grassi L, Nanni MG, Rodin G, Li M, Caruso R. The use of antidepressants in oncology: a review and practical tips for oncologists. Ann Oncol. 2018;29(1):101-111. doi:10.1093/annonc/mdx526
- Lee E, Park Y, Li D, Rodriguez-Fuguet A, Wang X, Zhang WC. Antidepressant use and lung cancer risk and survival: a meta-analysis of observational studies. Cancer Res Commun. 2023;3(6):1013-1025. doi:10.1158/2767-9764.CRC-23-0003
- Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Arch Gen Psychiatry. 2009;66(8):848 -856. doi:10.1001/archgenpsychiatry.2009.81
- Grattan A, Sullivan MD, Saunders KW, Campbell CI, Von Korff MR. Depression and prescription opioid misuse among chronic opioid therapy recipients with no history of substance abuse. Ann Fam Med. 2012;10(4):304-311. doi:10.1370/afm.1371
- Cowan DT, Wilson-Barnett J, Griffiths P, Allan LG. A survey of chronic noncancer pain patients prescribed opioid analgesics. Pain Med. 2003;4(4):340-351. doi:10.1111/j.1526-4637.2003.03038.x
- Breckenridge J, Clark JD. Patient characteristics associated with opioid versus nonsteroidal anti-inflammatory drug management of chronic low back pain. J Pain. 2003;4(6):344-350. doi:10.1016/s1526-5900(03)00638-2
- Edlund MJ, Martin BC, Devries A, Fan MY, Braden JB, Sullivan MD. Trends in use of opioids for chronic noncancer pain among individuals with mental health and substance use disorders: the TROUP study. Clin J Pain. 2010;26(1):1-8. doi:10.1097/AJP.0b013e3181b99f35
- Sullivan MD, Edlund MJ, Fan MY, DeVries A, Braden JB, Martin BC. Risks for possible and probable opioid misuse among recipients of chronic opioid therapy in commercial and medicaid insurance plans: the TROUP study. Pain. 2010;150(2):332-339. doi:10.1016/j.pain.2010.05.020
- Dunn KM, Saunders KW, Rutter CM, et al. Opioid prescriptions for chronic pain and overdose: a cohort study. Ann Intern Med. 2010;152(2):85-92. doi:10.7326/0003-4819-152-2-201001190-00006
- Haddad P. Do antidepressants have any potential to cause addiction? J Psychopharmacol. 1999;13(3):300- 307. doi:10.1177/026988119901300321
- Lakeview Health Staff. America’s most abused antidepressants. Lakeview Health. January 24, 2004. Accessed April 4, 2025. https://www.lakeviewhealth.com/blog/us-most-abused-antidepressants/
- Greenhouse Treatment Center Editorial Staff. Addiction to antidepressants: is it possible? America Addiction Centers: Greenhouse Treatment Center. Updated April 23, 2024. Accessed April 4, 2025. https://greenhousetreatment.com/prescription-medication/antidepressants/
Cancer patients experience depression at rates > 5 times that of the general population.1-11 Despite an increase in palliative care use, depression rates continued to rise.2-4 Between 5% to 16% of outpatients, 4% to 14% of inpatients, and up to 49% of patients receiving palliative care experience depression.5 This issue also impacts families and caregivers.1 A 2021 meta-analysis found that 23% of active military personnel and 20% of veterans experience depression.11
Antidepressants approved by the US Food and Drug Administration (FDA) target the serotonin, norepinephrine, or dopamine systems and include boxed warnings about an increased risk of suicidal thoughts in adults aged 18 to 24 years.12,13 These medications are categorized into several classes: monoamine oxidase inhibitors (MAOIs), tricyclic antidepressants (TCAs), tetracyclic antidepressants (TeCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), serotonin receptor modulators (SRMs), serotonin-melatonin receptor antagonists (SMRAs), and N—methyl-D-aspartate receptor antagonists (NMDARAs).14,15 The first FDA-approved antidepressants, iproniazid (an MAOI) and imipramine (a TCA) laid the foundation for the development of newer classes like SSRIs and SNRIs.15-17
Older antidepressants such as MAOIs and TCAs are used less due to their adverse effects (AEs) and drug interactions. MAOIs, such as iproniazid, selegiline, moclobemide, tranylcypromine, isocarboxazid, and phenelzine, have numerous AEs and drug interactions, making them unsuitable for first- or second-line treatment of depression.14,18-21 TCAs such as doxepin, amitriptyline, nortriptyline, imipramine, desipramine, clomipramine, trimipramine, protriptyline, maprotiline, and amoxapine have a narrow therapeutic index requiring careful monitoring for signs of toxicity such as QRS widening, tremors, or confusion. Despite the issues, TCAs are generally classified as second-line agents for major depressive disorder (MDD). TCAs have off-label uses for migraine prophylaxis, treatment of obsessive-compulsive disorder (OCD), insomnia, and chronic pain management first-line.14,22-29
Newer antidepressants, including TeCAs and NDRIs, are typically more effective, but also come with safety concerns. TeCAs like mirtazapine interact with several medications, including MAOIs, serotonin-increasing drugs, alcohol, cannabidiol, and marijuana. Mirtazapine is FDA-approved for the treatment of moderate to severe depression in adults. It is also used off-label to treat insomnia, panic disorder, posttraumatic stress disorder (PTSD), generalized anxiety disorder (GAD), social anxiety disorder (SAD), headaches, and migraines. Compared to other antidepressants, mirtazapine is effective for all stages of depression and addresses a broad range of related symptoms.14,30-34 NDRIs, such as bupropion, also interact with various medications, including MAOIs, other antidepressants, stimulants, and alcohol. Bupropion is FDA-approved for smoking cessation and to treat depression and SAD. It is also used off-label for depression- related bipolar disorder or sexual dysfunction, attention-deficit/hyperactivity disorder (ADHD), and obesity.14,35-42
SSRIs, SNRIs, and SRMs should be used with caution. SSRIs such as sertraline, citalopram, escitalopram, fluoxetine, paroxetine, and fluvoxamine are first-line treatments for depression and various psychiatric disorders due to their safety and efficacy. Common AEs of SSRIs include sexual dysfunction, sleep disturbances, weight changes, and gastrointestinal (GI) issues. SSRIs can prolong the QT interval, posing a risk of life-threatening arrhythmia, and may interact with other medications, necessitating treatment adjustments. The FDA approved SSRIs for MDD, GAD, bulimia nervosa, bipolar depression, OCD, panic disorder, premenstrual dysphoric disorder, treatment-resistant depression, PTSD, and SAD. Off-label uses include binge eating disorder, body dysmorphic disorder, fibromyalgia, premature ejaculation, paraphilias, autism, Raynaud phenomenon, and vasomotor symptoms associated with menopause. Among SSRIs, sertraline and escitalopram are noted for their effectiveness and tolerability.14,43-53
SNRIs, including duloxetine, venlafaxine, desvenlafaxine, milnacipran, and levomilnacipran, may increase bleeding risk, especially when taken with blood thinners. They can also elevate blood pressure, which may worsen if combined with stimulants. SNRIs may interact with other medications that affect serotonin levels, increasing the risk of serotonin syndrome when taken with triptans, pain medications, or other antidepressants.14 Desvenlafaxine has been approved by the FDA (but not by the European Medicines Agency).54-56 Duloxetine is FDA-approved for the treatment of depression, neuropathic pain, anxiety disorders, fibromyalgia, and musculoskeletal disorders. It is used off-label to treat chemotherapy-induced peripheral neuropathy and stress urinary incontinence.57-61 Venlafaxine is FDA-approved for depression, SAD, and panic disorder, and is prescribed off-label to treat ADHD, neuropathy, fibromyalgia, cataplexy, and PTSD, either alone or in combination with other medications.62,63 Milnacipran is not approved for MDD; levomilnacipran received approval in 2013.64
SRMs such as trazodone, nefazodone, vilazodone, and vortioxetine also function as serotonin reuptake inhibitors.14,15 Trazodone is FDA-approved for MDD. It has been used off-label to treat anxiety, Alzheimer disease, substance misuse, bulimia nervosa, insomnia, fibromyalgia, and PTSD when first-line SSRIs are ineffective. A notable AE of trazodone is orthostatic hypotension, which can lead to dizziness and increase the risk of falls, especially in geriatric patients.65-70 Nefazodone was discontinued in Europe in 2003 due to rare cases of liver toxicity but remains available in the US.71-74 Vilazodone and vortioxetine are FDA-approved.
The latest classes of antidepressants include SMRAs and NMDARAs.14 Agomelatine, an SMRA, was approved in Europe in 2009 but rejected by the FDA in 2011 due to liver toxicity.75 NMDARAs like esketamine and a combination of dextromethorphan and bupropion received FDA approval in 2019 and 2022, respectively.76,77
This retrospective study analyzes noncancer drugs used during systemic chemotherapy based on a dataset of 14 antineoplastic agents. It sought to identify the most dispensed noncancer drug groups, discuss findings, compare patients with and without antidepressant prescriptions, and examine trends in antidepressant use from 2002 to 2023. This analysis expands on prior research.78-81
Methods
The Walter Reed National Military Medical Center Institutional Review Board approved the study protocol and ensured compliance with the Health Insurance Portability and Accountability Act as an exempt protocol. The Joint Pathology Center (JPC) of the US Department of Defense (DoD) Cancer Registry Program and Military Health System (MHS) data experts from the Comprehensive Ambulatory/Professional Encounter Record (CAPER) and Pharmacy Data Transaction Service (PDTS) provided data for the analysis.
Data Sources
The JPC DoD Cancer Registry Program contains data from 1998 to 2024. CAPER and PDTS are part of the MHS Data Repository/Management Analysis and Reporting Tool database. Each observation in CAPER represents an ambulatory encounter at a military treatment facility (MTF). CAPER records are available from 2003 to 2024. PDTS records are available from 2002 to 2004. Each observation in PDTS represents a prescription filled for an MHS beneficiary, excluding those filled at international civilian pharmacies and inpatient pharmacy prescriptions.
This cross-sectional analysis requested data extraction for specific cancer drugs from the DoD Cancer Registry, focusing on treatment details, diagnosis dates, patient demographics, and physicians’ comments on AEs. After identifying patients, CAPER was used to identify additional health conditions. PDTS was used to compile a list of prescription medications filled during systemic cancer treatment or < 2 years postdiagnosis.
The 2016 Surveillance, Epidemiology, and End Results Program Coding and Staging Manual and International Classification of Diseases for Oncology, 3rd edition, 1st revision, were used to decode disease and cancer types.82,83 Data sorting and analysis were performed using Microsoft Excel. The percentage for the total was calculated by using the number of patients or data available within the subgroup divided by the total number of patients or data variables. To compare the mean number of dispensed antidepressants to those without antidepressants, a 2-tailed, 2-sample z test was used to calculate the P value and determine statistical significance (P < .05) using socscistatistics.com.
Data were extracted 3 times between 2021 and 2023. The initial 2021 protocol focused on erlotinib and gefitinib. A modified protocol in 2022 added paclitaxel, cisplatin, docetaxel, pemetrexed, and crizotinib; further modification in 2023 included 8 new antineoplastic agents and 2 anticoagulants. Sotorasib has not been prescribed in the MHS, and JPC lacks records for noncancer drugs. The 2023 dataset comprised 2210 patients with cancer treated with 14 antineoplastic agents; 2104 had documented diagnoses and 2113 had recorded prescriptions. Data for erlotinib, gefitinib, and paclitaxel have been published previously.78,79
Results
Of 2113 patients with recorded prescriptions, 1297 patients (61.4%) received 109 cancer drugs, including 96 antineoplastics, 7 disease-modifying antirheumatic agents, 4 biologic response modifiers, and 2 calcitonin gene-related peptides. Fourteen antineoplastic agents had complete data from JPC, while others were noted for combination therapies or treatment switches from the PDTS (Table 1). Seventy-six cancer drugs were prescribed with antidepressants in 489 patients (eAppendix).

The JPC provided 2242 entries for 2210 patients, ranging in age from 2 months to 88 years (mean, 56 years), documenting treatment from September 1988 to January 2023. Thirty-two patients had duplicate entries due to multiple cancer locations or occurrences. Of the 2242 patients, 1541 (68.7%) were aged > 50 years, 975 patients (43.5%) had cancers that were stage III or IV, and 1267 (56.5%) had cancers that were stage 0, I, II, or not applicable/unknown. There were 51 different types of cancer: breast, lung, testicular, endometrial, and ovarian were most common (n ≥ 100 patients). Forty-two cancer types were documented among 750 patients prescribed antidepressants (Table 2).

The CAPER database recorded 8882 unique diagnoses for 2104 patients, while PDTS noted 1089 unique prescriptions within 273 therapeutic codes for 2113 patients. Nine therapeutic codes (opiate agonists, adrenals, cathartics-laxatives, nonsteroidal anti-inflammatory agents, antihistamines for GI conditions, 5-HT3 receptor antagonists, analgesics and antipyretic miscellanea, antineoplastic agents, and proton-pump inhibitors) and 8 drugs (dexamethasone, prochlorperazine, ondansetron, docusate, acetaminophen, ibuprofen, oxycodone, and polyethylene glycol 3350) were associated with > 1000 patients (≥ 50%). Patients had between 1 and 275 unique health conditions and filled 1 to 108 prescriptions. The mean (SD) number of diagnoses and prescriptions was 50 (28) and 29 (12), respectively. Of the 273 therapeutic codes, 30 groups were analyzed, with others categorized into miscellaneous groups such as lotions, vaccines, and devices. Significant differences in mean number of prescriptions were found for patients taking antidepressants compared to those not (P < .05), except for anticonvulsants and antipsychotics (P = .12 and .09, respectively) (Table 3).

Antidepressants
Of the 2113 patients with recorded prescriptions, 750 (35.5%) were dispensed 17 different antidepressants. Among these 17 antidepressants, 183 (8.7%) patients received duloxetine, 158 (7.5%) received venlafaxine, 118 (5.6%) received trazodone, and 107 (5.1%) received sertraline (Figure 1, Table 4). Of the 750 patients, 509 (67.9%) received 1 antidepressant, 168 (22.4%) received 2, 60 (8.0%) received 3, and 13 (1.7%) received > 3. Combinations varied, but only duloxetine and trazodone were prescribed to > 10 patients.



Antidepressants were prescribed annually at an overall mean (SD) rate of 23% (5%) from 2003 to 2022 (Figure 2). Patients on antidepressants during systemic therapy had a greater number of diagnosed medical conditions and received more prescription medications compared to those not taking antidepressants (P < .001) (Figure 3). The 745 patients taking antidepressants in CAPER data had between 1 and 275 diagnosed medical issues, with a mean (SD) of 55 (31) vs a range of 1 to 209 and a mean (SD) of 46 (26) for the 1359 patients not taking antidepressants. The 750 patients on antidepressants in PDTS data had between 8 and 108 prescriptions dispensed, with a mean (SD) of 32 (12), vs a range of 1 to 65 prescriptions and a mean (SD) of 29 (12) for 1363 patients not taking antidepressants.


Discussion
The JPC DoD Cancer Registry includes information on cancer types, stages, treatment regimens, and physicians’ notes, while noncancer drugs are sourced from the PDTS database. The pharmacy uses a different documentation system, leading to varied classifications.
Database reliance has its drawbacks. For example, megestrol is coded as a cancer drug, although it’s primarily used for endometrial or gynecologic cancers. Many drugs have multiple therapeutic codes assigned to them, including 10 antineoplastic agents: diclofenac, Bacillus Calmette-Guérin (BCG), megestrol acetate, tamoxifen, anastrozole, letrozole, leuprolide, goserelin, degarelix, and fluorouracil. Diclofenac, BCG, and mitomycin have been repurposed for cancer treatment.84-87 From 2003 to 2023, diclofenac was prescribed to 350 patients for mild-to-moderate pain, with only 2 patients receiving it for cancer in 2018. FDA-approved for bladder cancer in 1990, BCG was prescribed for cancer treatment for 1 patient in 2021 after being used for vaccines between 2003 and 2018. Tamoxifen, used for hormone receptor-positive breast cancer from 2004 to 2017 with 53 patients, switched to estrogen agonist-antagonists from 2017 to 2023 with 123 patients. Only a few of the 168 patients were prescribed tamoxifen using both codes.88-91 Anastrozole and letrozole were coded as antiestrogens for 7 and 18 patients, respectively, while leuprolide and goserelin were coded as gonadotropins for 59 and 18 patients. Degarelix was coded as antigonadotropins, fluorouracil as skin and mucous membrane agents miscellaneous, and megestrol acetate as progestins for 7, 6, and 3 patients, respectively. Duloxetine was given to 186 patients, primarily for depression from 2005 to 2023, with 7 patients treated for fibromyalgia from 2022 to 2023.
Antidepressants Observed
Tables 1 and 5 provide insight into the FDA approval of 14 antineoplastics and antidepressants and their CYP metabolic pathways.92-122 In Table 4, the most prescribed antidepressant classes are SNRIs, SRMs, SSRIs, TeCAs, NDRIs, and TCAs. This trend highlights a preference for newer medications with weak CYP inhibition. A total of 349 patients were prescribed SSRIs, 343 SNRIs, 119 SRMs, 109 TCAs, 83 TeCAs, and 79 NDRIs. MAOIs, SMRAs, and NMDARAs were not observed in this dataset. While there are instances of dextromethorphan-bupropion and sertraline-escitalopram being dispensed together, it remains unclear whether these were NMDARA combinations.
Among the 14 specific antineoplastic agents, 10 are metabolized by CYP isoenzymes, primarily CYP3A4. Duloxetine neither inhibits nor is metabolized by CYP3A4, a reason it is often recommended, following venlafaxine.
Both duloxetine and venlafaxine are used off-label for chemotherapy-induced peripheral neuropathy related to paclitaxel and docetaxel. According to the CYP metabolized pathway, duloxetine tends to have more favorable DDIs than venlafaxine. In PDTS data, 371 patients were treated with paclitaxel and 180 with docetaxel, with respective antidepressant prescriptions of 156 and 70. Of the 156 patients dispensed paclitaxel, 62 (40%) were dispensed with duloxetine compared to 43 (28%) with venlafaxine. Of the 70 patients dispensed docetaxel, 23 (33%) received duloxetine vs 24 (34%) with venlafaxine.
Of 85 patients prescribed duloxetine, 75 received it with either paclitaxel or docetaxel (5 received both). Five patients had documented AEs (1 neuropathy related). Of 67 patients prescribed venlafaxine, 66 received it with either paclitaxel or docetaxel. Two patients had documented AEs (1 was neuropathy related, the same patient who received duloxetine). Of the 687 patients treated with paclitaxel and 337 with docetaxel in all databases, 4 experienced neuropathic AEs from both medications.79
Antidepressants can increase the risk of bleeding, especially when combined with blood thinners, and may elevate blood pressure, particularly alongside stimulants. Of the 554 patients prescribed 9 different anticoagulants, enoxaparin, apixaban, and rivaroxaban were the most common (each > 100 patients). Among these, 201 patients (36%) received both anticoagulants and antidepressants: duloxetine for 64 patients, venlafaxine for 30, trazodone for 35, and sertraline for 26. There were no data available to assess bleeding rates related to the evaluation of DDIs between these medication classes.
Antidepressants can be prescribed for erectile dysfunction. Of the 148 patients prescribed an antidepressant for erectile dysfunction, duloxetine, trazodone, and mirtazapine were the most common. Antidepressant preferences varied by cancer type. Duloxetine was the only antidepressant used for all types of cancer. Venlafaxine, duloxetine, trazodone, sertraline, and escitalopram were the most prescribed antidepressants for breast cancer, while duloxetine, mirtazapine, citalopram, sertraline, and trazodone were the most prescribed for lung cancer. Sertraline, duloxetine, trazodone, amitriptyline, and escitalopram were most common for testicular cancer. Duloxetine, venlafaxine, trazodone, amitriptyline, and sertraline were the most prescribed for endometrial cancer, while duloxetine, venlafaxine, amitriptyline, citalopram, and sertraline were most prescribed for ovarian cancer.
The broadness of International Statistical Classification of Diseases, Tenth Revision codes made it challenging to identify nondepression diagnoses in the analyzed population. However, if all antidepressants were prescribed to treat depression, service members with cancer exhibited a higher depression rate (35%) than the general population (25%). Of 2104 patients, 191 (9.1%) had mood disorders, and 706 (33.6%) had mental disorders: 346 (49.0%) had 1 diagnosis, and 360 (51.0%) had multiple diagnoses. The percentage of diagnoses varied yearly, with notable drops in 2003, 2007, 2011, 2014, and 2018, and peaks in 2006, 2008, 2013, 2017, and 2022. This fluctuation was influenced by events like the establishment of PDTS in 2002, the 2008 economic recession, a hospital relocation in 2011, the 2014 Ebola outbreak, and the COVID-19 pandemic. Although the number of patients receiving antidepressants increased from 2019 to 2022, the overall percentage of patients receiving them did not significantly change from 2003 to 2022, aligning with previous research.5,125
Many medications have potential uses beyond what is detailed in the prescribing information. Antidepressants can relieve pain, while pain medications may help with depression. Opioids were once thought to effectively treat depression, but this perspective has changed with a greater understanding of their risks, including misuse.126-131 Pain is a severe and often unbearable AE of cancer. Of 2113 patients, 92% received opioids; 34% received both opioids and antidepressants; 2% received only antidepressants; and 7% received neither. This study didn’t clarify whether those on opioids alone recognized their depression or if those on both were aware of their dependence. While SSRIs are generally not addictive, they can lead to physical dependence, and any medication can be abused if not managed properly.132-134
Conclusions
This retrospective study analyzes data from antineoplastic agents used in systemic cancer treatment between 1988 and 2023, with a particular focus on the use of antidepressants. Data on antidepressant prescriptions are incomplete and specific to these agents, which means the findings cannot be generalized to all antidepressants. Hence, the results indicate that patients taking antidepressants had more diagnosed health issues and received more medications compared to patients who were not on these drugs.
This study underscores the need for further research into the effects of antidepressants on cancer treatment, utilizing all data from the DoD Cancer Registry. Future research should explore DDIs between antidepressants and other cancer and noncancer medications, as this study did not assess AE documentation, unlike in studies involving erlotinib, gefitinib, and paclitaxel.78,79 Further investigation is needed to evaluate the impact of discontinuing antidepressant use during cancer treatment. This comprehensive overview provides insights for clinicians to help them make informed decisions regarding the prescription of antidepressants in the context of cancer treatment.
Cancer patients experience depression at rates > 5 times that of the general population.1-11 Despite an increase in palliative care use, depression rates continued to rise.2-4 Between 5% to 16% of outpatients, 4% to 14% of inpatients, and up to 49% of patients receiving palliative care experience depression.5 This issue also impacts families and caregivers.1 A 2021 meta-analysis found that 23% of active military personnel and 20% of veterans experience depression.11
Antidepressants approved by the US Food and Drug Administration (FDA) target the serotonin, norepinephrine, or dopamine systems and include boxed warnings about an increased risk of suicidal thoughts in adults aged 18 to 24 years.12,13 These medications are categorized into several classes: monoamine oxidase inhibitors (MAOIs), tricyclic antidepressants (TCAs), tetracyclic antidepressants (TeCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), serotonin receptor modulators (SRMs), serotonin-melatonin receptor antagonists (SMRAs), and N—methyl-D-aspartate receptor antagonists (NMDARAs).14,15 The first FDA-approved antidepressants, iproniazid (an MAOI) and imipramine (a TCA) laid the foundation for the development of newer classes like SSRIs and SNRIs.15-17
Older antidepressants such as MAOIs and TCAs are used less due to their adverse effects (AEs) and drug interactions. MAOIs, such as iproniazid, selegiline, moclobemide, tranylcypromine, isocarboxazid, and phenelzine, have numerous AEs and drug interactions, making them unsuitable for first- or second-line treatment of depression.14,18-21 TCAs such as doxepin, amitriptyline, nortriptyline, imipramine, desipramine, clomipramine, trimipramine, protriptyline, maprotiline, and amoxapine have a narrow therapeutic index requiring careful monitoring for signs of toxicity such as QRS widening, tremors, or confusion. Despite the issues, TCAs are generally classified as second-line agents for major depressive disorder (MDD). TCAs have off-label uses for migraine prophylaxis, treatment of obsessive-compulsive disorder (OCD), insomnia, and chronic pain management first-line.14,22-29
Newer antidepressants, including TeCAs and NDRIs, are typically more effective, but also come with safety concerns. TeCAs like mirtazapine interact with several medications, including MAOIs, serotonin-increasing drugs, alcohol, cannabidiol, and marijuana. Mirtazapine is FDA-approved for the treatment of moderate to severe depression in adults. It is also used off-label to treat insomnia, panic disorder, posttraumatic stress disorder (PTSD), generalized anxiety disorder (GAD), social anxiety disorder (SAD), headaches, and migraines. Compared to other antidepressants, mirtazapine is effective for all stages of depression and addresses a broad range of related symptoms.14,30-34 NDRIs, such as bupropion, also interact with various medications, including MAOIs, other antidepressants, stimulants, and alcohol. Bupropion is FDA-approved for smoking cessation and to treat depression and SAD. It is also used off-label for depression- related bipolar disorder or sexual dysfunction, attention-deficit/hyperactivity disorder (ADHD), and obesity.14,35-42
SSRIs, SNRIs, and SRMs should be used with caution. SSRIs such as sertraline, citalopram, escitalopram, fluoxetine, paroxetine, and fluvoxamine are first-line treatments for depression and various psychiatric disorders due to their safety and efficacy. Common AEs of SSRIs include sexual dysfunction, sleep disturbances, weight changes, and gastrointestinal (GI) issues. SSRIs can prolong the QT interval, posing a risk of life-threatening arrhythmia, and may interact with other medications, necessitating treatment adjustments. The FDA approved SSRIs for MDD, GAD, bulimia nervosa, bipolar depression, OCD, panic disorder, premenstrual dysphoric disorder, treatment-resistant depression, PTSD, and SAD. Off-label uses include binge eating disorder, body dysmorphic disorder, fibromyalgia, premature ejaculation, paraphilias, autism, Raynaud phenomenon, and vasomotor symptoms associated with menopause. Among SSRIs, sertraline and escitalopram are noted for their effectiveness and tolerability.14,43-53
SNRIs, including duloxetine, venlafaxine, desvenlafaxine, milnacipran, and levomilnacipran, may increase bleeding risk, especially when taken with blood thinners. They can also elevate blood pressure, which may worsen if combined with stimulants. SNRIs may interact with other medications that affect serotonin levels, increasing the risk of serotonin syndrome when taken with triptans, pain medications, or other antidepressants.14 Desvenlafaxine has been approved by the FDA (but not by the European Medicines Agency).54-56 Duloxetine is FDA-approved for the treatment of depression, neuropathic pain, anxiety disorders, fibromyalgia, and musculoskeletal disorders. It is used off-label to treat chemotherapy-induced peripheral neuropathy and stress urinary incontinence.57-61 Venlafaxine is FDA-approved for depression, SAD, and panic disorder, and is prescribed off-label to treat ADHD, neuropathy, fibromyalgia, cataplexy, and PTSD, either alone or in combination with other medications.62,63 Milnacipran is not approved for MDD; levomilnacipran received approval in 2013.64
SRMs such as trazodone, nefazodone, vilazodone, and vortioxetine also function as serotonin reuptake inhibitors.14,15 Trazodone is FDA-approved for MDD. It has been used off-label to treat anxiety, Alzheimer disease, substance misuse, bulimia nervosa, insomnia, fibromyalgia, and PTSD when first-line SSRIs are ineffective. A notable AE of trazodone is orthostatic hypotension, which can lead to dizziness and increase the risk of falls, especially in geriatric patients.65-70 Nefazodone was discontinued in Europe in 2003 due to rare cases of liver toxicity but remains available in the US.71-74 Vilazodone and vortioxetine are FDA-approved.
The latest classes of antidepressants include SMRAs and NMDARAs.14 Agomelatine, an SMRA, was approved in Europe in 2009 but rejected by the FDA in 2011 due to liver toxicity.75 NMDARAs like esketamine and a combination of dextromethorphan and bupropion received FDA approval in 2019 and 2022, respectively.76,77
This retrospective study analyzes noncancer drugs used during systemic chemotherapy based on a dataset of 14 antineoplastic agents. It sought to identify the most dispensed noncancer drug groups, discuss findings, compare patients with and without antidepressant prescriptions, and examine trends in antidepressant use from 2002 to 2023. This analysis expands on prior research.78-81
Methods
The Walter Reed National Military Medical Center Institutional Review Board approved the study protocol and ensured compliance with the Health Insurance Portability and Accountability Act as an exempt protocol. The Joint Pathology Center (JPC) of the US Department of Defense (DoD) Cancer Registry Program and Military Health System (MHS) data experts from the Comprehensive Ambulatory/Professional Encounter Record (CAPER) and Pharmacy Data Transaction Service (PDTS) provided data for the analysis.
Data Sources
The JPC DoD Cancer Registry Program contains data from 1998 to 2024. CAPER and PDTS are part of the MHS Data Repository/Management Analysis and Reporting Tool database. Each observation in CAPER represents an ambulatory encounter at a military treatment facility (MTF). CAPER records are available from 2003 to 2024. PDTS records are available from 2002 to 2004. Each observation in PDTS represents a prescription filled for an MHS beneficiary, excluding those filled at international civilian pharmacies and inpatient pharmacy prescriptions.
This cross-sectional analysis requested data extraction for specific cancer drugs from the DoD Cancer Registry, focusing on treatment details, diagnosis dates, patient demographics, and physicians’ comments on AEs. After identifying patients, CAPER was used to identify additional health conditions. PDTS was used to compile a list of prescription medications filled during systemic cancer treatment or < 2 years postdiagnosis.
The 2016 Surveillance, Epidemiology, and End Results Program Coding and Staging Manual and International Classification of Diseases for Oncology, 3rd edition, 1st revision, were used to decode disease and cancer types.82,83 Data sorting and analysis were performed using Microsoft Excel. The percentage for the total was calculated by using the number of patients or data available within the subgroup divided by the total number of patients or data variables. To compare the mean number of dispensed antidepressants to those without antidepressants, a 2-tailed, 2-sample z test was used to calculate the P value and determine statistical significance (P < .05) using socscistatistics.com.
Data were extracted 3 times between 2021 and 2023. The initial 2021 protocol focused on erlotinib and gefitinib. A modified protocol in 2022 added paclitaxel, cisplatin, docetaxel, pemetrexed, and crizotinib; further modification in 2023 included 8 new antineoplastic agents and 2 anticoagulants. Sotorasib has not been prescribed in the MHS, and JPC lacks records for noncancer drugs. The 2023 dataset comprised 2210 patients with cancer treated with 14 antineoplastic agents; 2104 had documented diagnoses and 2113 had recorded prescriptions. Data for erlotinib, gefitinib, and paclitaxel have been published previously.78,79
Results
Of 2113 patients with recorded prescriptions, 1297 patients (61.4%) received 109 cancer drugs, including 96 antineoplastics, 7 disease-modifying antirheumatic agents, 4 biologic response modifiers, and 2 calcitonin gene-related peptides. Fourteen antineoplastic agents had complete data from JPC, while others were noted for combination therapies or treatment switches from the PDTS (Table 1). Seventy-six cancer drugs were prescribed with antidepressants in 489 patients (eAppendix).

The JPC provided 2242 entries for 2210 patients, ranging in age from 2 months to 88 years (mean, 56 years), documenting treatment from September 1988 to January 2023. Thirty-two patients had duplicate entries due to multiple cancer locations or occurrences. Of the 2242 patients, 1541 (68.7%) were aged > 50 years, 975 patients (43.5%) had cancers that were stage III or IV, and 1267 (56.5%) had cancers that were stage 0, I, II, or not applicable/unknown. There were 51 different types of cancer: breast, lung, testicular, endometrial, and ovarian were most common (n ≥ 100 patients). Forty-two cancer types were documented among 750 patients prescribed antidepressants (Table 2).

The CAPER database recorded 8882 unique diagnoses for 2104 patients, while PDTS noted 1089 unique prescriptions within 273 therapeutic codes for 2113 patients. Nine therapeutic codes (opiate agonists, adrenals, cathartics-laxatives, nonsteroidal anti-inflammatory agents, antihistamines for GI conditions, 5-HT3 receptor antagonists, analgesics and antipyretic miscellanea, antineoplastic agents, and proton-pump inhibitors) and 8 drugs (dexamethasone, prochlorperazine, ondansetron, docusate, acetaminophen, ibuprofen, oxycodone, and polyethylene glycol 3350) were associated with > 1000 patients (≥ 50%). Patients had between 1 and 275 unique health conditions and filled 1 to 108 prescriptions. The mean (SD) number of diagnoses and prescriptions was 50 (28) and 29 (12), respectively. Of the 273 therapeutic codes, 30 groups were analyzed, with others categorized into miscellaneous groups such as lotions, vaccines, and devices. Significant differences in mean number of prescriptions were found for patients taking antidepressants compared to those not (P < .05), except for anticonvulsants and antipsychotics (P = .12 and .09, respectively) (Table 3).

Antidepressants
Of the 2113 patients with recorded prescriptions, 750 (35.5%) were dispensed 17 different antidepressants. Among these 17 antidepressants, 183 (8.7%) patients received duloxetine, 158 (7.5%) received venlafaxine, 118 (5.6%) received trazodone, and 107 (5.1%) received sertraline (Figure 1, Table 4). Of the 750 patients, 509 (67.9%) received 1 antidepressant, 168 (22.4%) received 2, 60 (8.0%) received 3, and 13 (1.7%) received > 3. Combinations varied, but only duloxetine and trazodone were prescribed to > 10 patients.



Antidepressants were prescribed annually at an overall mean (SD) rate of 23% (5%) from 2003 to 2022 (Figure 2). Patients on antidepressants during systemic therapy had a greater number of diagnosed medical conditions and received more prescription medications compared to those not taking antidepressants (P < .001) (Figure 3). The 745 patients taking antidepressants in CAPER data had between 1 and 275 diagnosed medical issues, with a mean (SD) of 55 (31) vs a range of 1 to 209 and a mean (SD) of 46 (26) for the 1359 patients not taking antidepressants. The 750 patients on antidepressants in PDTS data had between 8 and 108 prescriptions dispensed, with a mean (SD) of 32 (12), vs a range of 1 to 65 prescriptions and a mean (SD) of 29 (12) for 1363 patients not taking antidepressants.


Discussion
The JPC DoD Cancer Registry includes information on cancer types, stages, treatment regimens, and physicians’ notes, while noncancer drugs are sourced from the PDTS database. The pharmacy uses a different documentation system, leading to varied classifications.
Database reliance has its drawbacks. For example, megestrol is coded as a cancer drug, although it’s primarily used for endometrial or gynecologic cancers. Many drugs have multiple therapeutic codes assigned to them, including 10 antineoplastic agents: diclofenac, Bacillus Calmette-Guérin (BCG), megestrol acetate, tamoxifen, anastrozole, letrozole, leuprolide, goserelin, degarelix, and fluorouracil. Diclofenac, BCG, and mitomycin have been repurposed for cancer treatment.84-87 From 2003 to 2023, diclofenac was prescribed to 350 patients for mild-to-moderate pain, with only 2 patients receiving it for cancer in 2018. FDA-approved for bladder cancer in 1990, BCG was prescribed for cancer treatment for 1 patient in 2021 after being used for vaccines between 2003 and 2018. Tamoxifen, used for hormone receptor-positive breast cancer from 2004 to 2017 with 53 patients, switched to estrogen agonist-antagonists from 2017 to 2023 with 123 patients. Only a few of the 168 patients were prescribed tamoxifen using both codes.88-91 Anastrozole and letrozole were coded as antiestrogens for 7 and 18 patients, respectively, while leuprolide and goserelin were coded as gonadotropins for 59 and 18 patients. Degarelix was coded as antigonadotropins, fluorouracil as skin and mucous membrane agents miscellaneous, and megestrol acetate as progestins for 7, 6, and 3 patients, respectively. Duloxetine was given to 186 patients, primarily for depression from 2005 to 2023, with 7 patients treated for fibromyalgia from 2022 to 2023.
Antidepressants Observed
Tables 1 and 5 provide insight into the FDA approval of 14 antineoplastics and antidepressants and their CYP metabolic pathways.92-122 In Table 4, the most prescribed antidepressant classes are SNRIs, SRMs, SSRIs, TeCAs, NDRIs, and TCAs. This trend highlights a preference for newer medications with weak CYP inhibition. A total of 349 patients were prescribed SSRIs, 343 SNRIs, 119 SRMs, 109 TCAs, 83 TeCAs, and 79 NDRIs. MAOIs, SMRAs, and NMDARAs were not observed in this dataset. While there are instances of dextromethorphan-bupropion and sertraline-escitalopram being dispensed together, it remains unclear whether these were NMDARA combinations.
Among the 14 specific antineoplastic agents, 10 are metabolized by CYP isoenzymes, primarily CYP3A4. Duloxetine neither inhibits nor is metabolized by CYP3A4, a reason it is often recommended, following venlafaxine.
Both duloxetine and venlafaxine are used off-label for chemotherapy-induced peripheral neuropathy related to paclitaxel and docetaxel. According to the CYP metabolized pathway, duloxetine tends to have more favorable DDIs than venlafaxine. In PDTS data, 371 patients were treated with paclitaxel and 180 with docetaxel, with respective antidepressant prescriptions of 156 and 70. Of the 156 patients dispensed paclitaxel, 62 (40%) were dispensed with duloxetine compared to 43 (28%) with venlafaxine. Of the 70 patients dispensed docetaxel, 23 (33%) received duloxetine vs 24 (34%) with venlafaxine.
Of 85 patients prescribed duloxetine, 75 received it with either paclitaxel or docetaxel (5 received both). Five patients had documented AEs (1 neuropathy related). Of 67 patients prescribed venlafaxine, 66 received it with either paclitaxel or docetaxel. Two patients had documented AEs (1 was neuropathy related, the same patient who received duloxetine). Of the 687 patients treated with paclitaxel and 337 with docetaxel in all databases, 4 experienced neuropathic AEs from both medications.79
Antidepressants can increase the risk of bleeding, especially when combined with blood thinners, and may elevate blood pressure, particularly alongside stimulants. Of the 554 patients prescribed 9 different anticoagulants, enoxaparin, apixaban, and rivaroxaban were the most common (each > 100 patients). Among these, 201 patients (36%) received both anticoagulants and antidepressants: duloxetine for 64 patients, venlafaxine for 30, trazodone for 35, and sertraline for 26. There were no data available to assess bleeding rates related to the evaluation of DDIs between these medication classes.
Antidepressants can be prescribed for erectile dysfunction. Of the 148 patients prescribed an antidepressant for erectile dysfunction, duloxetine, trazodone, and mirtazapine were the most common. Antidepressant preferences varied by cancer type. Duloxetine was the only antidepressant used for all types of cancer. Venlafaxine, duloxetine, trazodone, sertraline, and escitalopram were the most prescribed antidepressants for breast cancer, while duloxetine, mirtazapine, citalopram, sertraline, and trazodone were the most prescribed for lung cancer. Sertraline, duloxetine, trazodone, amitriptyline, and escitalopram were most common for testicular cancer. Duloxetine, venlafaxine, trazodone, amitriptyline, and sertraline were the most prescribed for endometrial cancer, while duloxetine, venlafaxine, amitriptyline, citalopram, and sertraline were most prescribed for ovarian cancer.
The broadness of International Statistical Classification of Diseases, Tenth Revision codes made it challenging to identify nondepression diagnoses in the analyzed population. However, if all antidepressants were prescribed to treat depression, service members with cancer exhibited a higher depression rate (35%) than the general population (25%). Of 2104 patients, 191 (9.1%) had mood disorders, and 706 (33.6%) had mental disorders: 346 (49.0%) had 1 diagnosis, and 360 (51.0%) had multiple diagnoses. The percentage of diagnoses varied yearly, with notable drops in 2003, 2007, 2011, 2014, and 2018, and peaks in 2006, 2008, 2013, 2017, and 2022. This fluctuation was influenced by events like the establishment of PDTS in 2002, the 2008 economic recession, a hospital relocation in 2011, the 2014 Ebola outbreak, and the COVID-19 pandemic. Although the number of patients receiving antidepressants increased from 2019 to 2022, the overall percentage of patients receiving them did not significantly change from 2003 to 2022, aligning with previous research.5,125
Many medications have potential uses beyond what is detailed in the prescribing information. Antidepressants can relieve pain, while pain medications may help with depression. Opioids were once thought to effectively treat depression, but this perspective has changed with a greater understanding of their risks, including misuse.126-131 Pain is a severe and often unbearable AE of cancer. Of 2113 patients, 92% received opioids; 34% received both opioids and antidepressants; 2% received only antidepressants; and 7% received neither. This study didn’t clarify whether those on opioids alone recognized their depression or if those on both were aware of their dependence. While SSRIs are generally not addictive, they can lead to physical dependence, and any medication can be abused if not managed properly.132-134
Conclusions
This retrospective study analyzes data from antineoplastic agents used in systemic cancer treatment between 1988 and 2023, with a particular focus on the use of antidepressants. Data on antidepressant prescriptions are incomplete and specific to these agents, which means the findings cannot be generalized to all antidepressants. Hence, the results indicate that patients taking antidepressants had more diagnosed health issues and received more medications compared to patients who were not on these drugs.
This study underscores the need for further research into the effects of antidepressants on cancer treatment, utilizing all data from the DoD Cancer Registry. Future research should explore DDIs between antidepressants and other cancer and noncancer medications, as this study did not assess AE documentation, unlike in studies involving erlotinib, gefitinib, and paclitaxel.78,79 Further investigation is needed to evaluate the impact of discontinuing antidepressant use during cancer treatment. This comprehensive overview provides insights for clinicians to help them make informed decisions regarding the prescription of antidepressants in the context of cancer treatment.
- National Cancer Institute. Depression (PDQ)-Health Professional Version. Updated July 25, 2024. Accessed April 4, 2025. https://www.cancer.gov/about-cancer/coping/feelings/depression-hp-pdq
- Krebber AM, Buffart LM, Kleijn G, et al. Prevalence of depression in cancer patients: a meta-analysis of diagnostic interviews and self-report instruments. Psychooncology. 2014;23(2):121-130. doi:10.1002/pon.3409
- Xiang X, An R, Gehlert S. Trends in antidepressant use among U.S. cancer survivors, 1999-2012. Psychiatr Serv. 2015;66(6):564. doi:10.1176/appi.ps.201500007
- Hartung TJ, Brähler E, Faller H, et al. The risk of being depressed is significantly higher in cancer patients than in the general population: Prevalence and severity of depressive symptoms across major cancer types. Eur J Cancer. 2017;72:46-53. doi:10.1016/j.ejca.2016.11.017
- Walker J, Holm Hansen C, Martin P, et al. Prevalence of depression in adults with cancer: a systematic review. Ann Oncol. 2013;24(4):895-900. doi:10.1093/annonc/mds575
- Pitman A, Suleman S, Hyde N, Hodgkiss A. Depression and anxiety in patients with cancer. BMJ. 2018;361:k1415. doi:10.1136/bmj.k1415
- Kisely S, Alotiby MKN, Protani MM, Soole R, Arnautovska U, Siskind D. Breast cancer treatment disparities in patients with severe mental illness: a systematic review and meta-analysis. Psychooncology. 2023;32(5):651-662. doi:10.1002/pon.6120
- Massie MJ. Prevalence of depression in patients with cancer. J Natl Cancer Inst Monogr. 2004;(32):57-71. doi:10.1093/jncimonographs/lgh014
- Rodin G, Katz M, Lloyd N, Green E, Mackay JA, Wong RK. Treatment of depression in cancer patients. Curr Oncol. 2007;14(5):180-188. doi:10.3747/co.2007.146
- Wilson KG, Chochinov HM, Skirko MG, et al. Depression and anxiety disorders in palliative cancer care. J Pain Symptom Manage. 2007;33(2):118-129. doi:10.1016/j.jpainsymman.2006.07.016
- Moradi Y, Dowran B, Sepandi M. The global prevalence of depression, suicide ideation, and attempts in the military forces: a systematic review and meta-analysis of cross sectional studies. BMC Psychiatry. 2021;21(1):510. doi:10.1186/s12888-021-03526-2
- Lu CY, Zhang F, Lakoma MD, et al. Changes in antidepressant use by young people and suicidal behavior after FDA warnings and media coverage: quasi-experimental study. BMJ. 2014;348:g3596. doi:10.1136/bmj.g3596
- Friedman RA. Antidepressants’ black-box warning--10 years later. N Engl J Med. 2014;371(18):1666-1668. doi:10.1056/NEJMp1408480
- Sheffler ZM, Patel P, Abdijadid S. Antidepressants. In: StatPearls. StatPearls Publishing. Updated May 26, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK538182/
- Miller JJ. Antidepressants, Part 1: 100 Years and Counting. Accessed April 4, 2025. Psychiatric Times. https:// www.psychiatrictimes.com/view/antidepressants-part-1-100-years-and-counting
- Hillhouse TM, Porter JH. A brief history of the development of antidepressant drugs: from monoamines to glutamate. Exp Clin Psychopharmacol. 2015;23(1):1-21. doi:10.1037/a0038550
- Mitchell PB, Mitchell MS. The management of depression. Part 2. The place of the new antidepressants. Aust Fam Physician. 1994;23(9):1771-1781.
- Sabri MA, Saber-Ayad MM. MAO Inhibitors. In: Stat- Pearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557395/
- Sub Laban T, Saadabadi A. Monoamine Oxidase Inhibitors (MAOI). In: StatPearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK539848/
- Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract. 2004;10(4):239-248. doi:10.1097/00131746-200407000-00005
- Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03
- Moraczewski J, Awosika AO, Aedma KK. Tricyclic Antidepressants. In: StatPearls. StatPearls Publishing. Updated August 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557791/
- Almasi A, Patel P, Meza CE. Doxepin. In: StatPearls. StatPearls Publishing. Updated February 14, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK542306/
- Thour A, Marwaha R. Amitriptyline. In: StatPearls. Stat- Pearls Publishing. Updated July 18, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK537225/
- Radley DC, Finkelstein SN, Stafford RS. Off-label prescribing among office-based physicians. Arch Intern Med. 2006;166(9):1021-1026. doi:10.1001/archinte.166.9.1021
- Tesfaye S, Sloan G, Petrie J, et al. Comparison of amitriptyline supplemented with pregabalin, pregabalin supplemented with amitriptyline, and duloxetine supplemented with pregabalin for the treatment of diabetic peripheral neuropathic pain (OPTION-DM): a multicentre, double-blind, randomised crossover trial. Lancet. 2022;400(10353):680- 690. doi:10.1016/S0140-6736(22)01472-6
- Farag HM, Yunusa I, Goswami H, Sultan I, Doucette JA, Eguale T. Comparison of amitriptyline and US Food and Drug Administration-approved treatments for fibromyalgia: a systematic review and network metaanalysis. JAMA Netw Open. 2022;5(5):e2212939. doi:10.1001/jamanetworkopen.2022.12939
- Merwar G, Gibbons JR, Hosseini SA, Saadabadi A. Nortriptyline. In: StatPearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482214/
- Fayez R, Gupta V. Imipramine. In: StatPearls. StatPearls Publishing. Updated May 22, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557656/
- Jilani TN, Gibbons JR, Faizy RM, Saadabadi A. Mirtazapine. In: StatPearls. StatPearls Publishing. Updated August 28, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK519059/
- Nutt DJ. Tolerability and safety aspects of mirtazapine. Hum Psychopharmacol. 2002;17 Suppl 1:S37-S41. doi:10.1002/hup.388
- Gandotra K, Chen P, Jaskiw GE, Konicki PE, Strohl KP. Effective treatment of insomnia with mirtazapine attenuates concomitant suicidal ideation. J Clin Sleep Med. 2018;14(5):901-902. doi:10.5664/jcsm.7142
- Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249-264. doi:10.1111/j.1527-3458.2001.tb00198.x
- Wang SM, Han C, Bahk WM, et al. Addressing the side effects of contemporary antidepressant drugs: a comprehensive review. Chonnam Med J. 2018;54(2):101-112. doi:10.4068/cmj.2018.54.2.101
- Huecker MR, Smiley A, Saadabadi A. Bupropion. In: StatPearls. StatPearls Publishing. Updated April 9, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470212/
- Hsieh MT, Tseng PT, Wu YC, et al. Effects of different pharmacologic smoking cessation treatments on body weight changes and success rates in patients with nicotine dependence: a network meta-analysis. Obes Rev. 2019;20(6):895- 905. doi:10.1111/obr.12835
- Hankosky ER, Bush HM, Dwoskin LP, et al. Retrospective analysis of health claims to evaluate pharmacotherapies with potential for repurposing: association of bupropion and stimulant use disorder remission. AMIA Annu Symp Proc. 2018;2018:1292-1299.
- Livingstone-Banks J, Norris E, Hartmann-Boyce J, West R, Jarvis M, Hajek P. Relapse prevention interventions for smoking cessation. Cochrane Database Syst Rev. 2019; 2(2):CD003999. doi:10.1002/14651858.CD003999.pub5
- Clayton AH, Kingsberg SA, Goldstein I. Evaluation and management of hypoactive sexual desire disorder. Sex Med. 2018;6(2):59-74. doi:10.1016/j.esxm.2018.01.004
- Verbeeck W, Bekkering GE, Van den Noortgate W, Kramers C. Bupropion for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev. 2017;10(10):CD009504. doi:10.1002/14651858.CD009504.pub2
- Ng QX. A systematic review of the use of bupropion for attention-deficit/hyperactivity disorder in children and adolescents. J Child Adolesc Psychopharmacol. 2017;27(2):112-116. doi:10.1089/cap.2016.0124
- Fava M, Rush AJ, Thase ME, et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim Care Companion J Clin Psychiatry. 2005;7(3):106-113. doi:10.4088/pcc.v07n0305
- Chu A, Wadhwa R. Selective Serotonin Reuptake Inhibitors. In: StatPearls. StatPearls Publishing. Updated May 1, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK554406/
- Singh HK, Saadabadi A. Sertraline. In: StatPearls. Stat- Pearls Publishing. Updated February 13, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK547689/
- MacQueen G, Born L, Steiner M. The selective serotonin reuptake inhibitor sertraline: its profile and use in psychiatric disorders. CNS Drug Rev. 2001;7(1):1-24. doi:10.1111/j.1527-3458.2001.tb00188.x
- Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatments meta-analysis. Lancet. 2009;373(9665):746-758. doi:10.1016/S0140-6736(09)60046-5
- Nelson JC. The STAR*D study: a four-course meal that leaves us wanting more. Am J Psychiatry. 2006;163(11):1864-1866. doi:10.1176/ajp.2006.163.11.1864
- Sharbaf Shoar N, Fariba KA, Padhy RK. Citalopram. In: StatPearls. StatPearls Publishing. Updated November 7, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482222/
- Landy K, Rosani A, Estevez R. Escitalopram. In: Stat- Pearls. StatPearls Publishing. Updated November 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557734/
- Cavanah LR, Ray P, Goldhirsh JL, Huey LY, Piper BJ. Rise of escitalopram and the fall of citalopram. medRxiv. Preprint published online May 8, 2023. doi:10.1101/2023.05.07.23289632
- Sohel AJ, Shutter MC, Patel P, Molla M. Fluoxetine. In: StatPearls. StatPearls Publishing. Updated July 4, 2022. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK459223/
- Wong DT, Perry KW, Bymaster FP. Case history: the discovery of fluoxetine hydrochloride (Prozac). Nat Rev Drug Discov. 2005;4(9):764-774. doi:10.1038/nrd1821
- Shrestha P, Fariba KA, Abdijadid S. Paroxetine. In: Stat- Pearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK526022/
- Naseeruddin R, Rosani A, Marwaha R. Desvenlafaxine. In: StatPearls. StatPearls Publishing. Updated July 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK534829
- Lieberman DZ, Massey SH. Desvenlafaxine in major depressive disorder: an evidence-based review of its place in therapy. Core Evid. 2010;4:67-82. doi:10.2147/ce.s5998
- Withdrawal assessment report for Ellefore (desvenlafaxine). European Medicine Agency. 2009. Accessed April 4, 2025. https://www.ema.europa.eu/en/documents/withdrawal-report/withdrawal-assessment-report-ellefore_en.pdf
- Dhaliwal JS, Spurling BC, Molla M. Duloxetine. In: Stat- Pearls. Statpearls Publishing. Updated May 29, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK549806/
- Hershman DL, Lacchetti C, Dworkin RH, et al. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2014;32(18):1941-1967. doi:10.1200/JCO.2013.54.0914
- Sommer C, Häuser W, Alten R, et al. Medikamentöse Therapie des Fibromyalgiesyndroms. Systematische Übersicht und Metaanalyse [Drug therapy of fibromyalgia syndrome. Systematic review, meta-analysis and guideline]. Schmerz. 2012;26(3):297-310. doi:10.1007/s00482-012-1172-2
- Bril V, England J, Franklin GM, et al. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology. 2011;76(20):1758-1765. doi:10.1212/WNL.0b013e3182166ebe
- Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2010;17(9):1113-e88. doi:10.1111/j.1468-1331.2010.02999.x
- Singh D, Saadabadi A. Venlafaxine. In: StatPearls. StatePearls Publishing. Updated February 26, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK535363/
- Sertraline and venlafaxine: new indication. Prevention of recurrent depression: no advance. Prescrire Int. 2005;14(75):19-20.
- Bruno A, Morabito P, S p i n a E , M u s c a t ello MRl. The role of levomilnacipran in the management of major depressive disorder: a comprehensive review. Curr Neuro pharmacol. 2016;14(2):191-199. doi:10.2174/1570159x14666151117122458
- Shin JJ, Saadabadi A. Trazodone. In: StatPearls. StatPearls Publishing. Updated February 29, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470560/
- Khouzam HR. A review of trazodone use in psychiatric and medical conditions. Postgrad Med. 2017;129(1):140-148. doi:10.1080/00325481.2017.1249265
- Smales ET, Edwards BA, Deyoung PN, et al. Trazodone effects on obstructive sleep apnea and non-REM arousal threshold. Ann Am Thorac Soc. 2015;12(5):758-764. doi:10.1513/AnnalsATS.201408-399OC
- Eckert DJ, Malhotra A, Wellman A, White DP. Trazodone increases the respiratory arousal threshold in patients with obstructive sleep apnea and a low arousal threshold. Sleep. 2014;37(4):811-819. doi:10.5665/sleep.3596
- Schatzberg AF, Nemeroff CB, eds. The American Psychiat ric Association Publishing Textbook of Psychopharmacology. 4th ed. American Psychiatric Publishing; 2009.
- Ruxton K, Woodman RJ, Mangoni AA. Drugs with anticholinergic effects and cognitive impairment, falls and allcause mortality in older adults: a systematic review and meta-analysis. Br J Clin Pharmacol. 2015;80(2):209-220. doi:10.1111/bcp.12617
- Nefazodone. In: LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. National Institute of Diabetes and Digestive and Kidney Diseases. Updated March 6, 2020. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK548179/
- Drugs of Current Interest: Nefazodone. WHO Pharmaceuticals Newsletter:(1). 2003(1):7. https://web.archive.org/web/20150403165029/http:/apps.who.int/medicinedocs/en/d/Js4944e/3.html
- Choi S. Nefazodone (serzone) withdrawn because of hepatotoxicity. CMAJ. 2003;169(11):1187.
- Teva Nefazodone Statement. News release. Teva USA. December 20, 2021. Accessed April 4, 2025. https:// www.tevausa.com/news-and-media/press-releases/teva-nefazodone-statement/
- Levitan MN, Papelbaum M, Nardi AE. Profile of agomelatine and its potential in the treatment of generalized anxiety disorder. Neuropsychiatr Dis Treat. 2015;11:1149- 1155. doi:10.2147/NDT.S67470
- Fu DJ, Ionescu DF, Li X, et al. Esketamine nasal spray for rapid reduction of major depressive disorder symptoms in patients who have active suicidal ideation with intent: double-blind, randomized study (ASPIRE I). J Clin Psychiatry. 2020;81(3):19m13191. doi:10.4088/JCP.19m13191
- Iosifescu DV, Jones A, O’Gorman C, et al. Efficacy and safety of AXS-05 (dextromethorphan-bupropion) in patients with major depressive disorder: a phase 3 randomized clinical trial (GEMINI). J Clin Psychiatry. 2022;83(4): 21m14345. doi:10.4088/JCP.21m14345
- Luong TT, Powers CN, Reinhardt BJ, et al. Retrospective evaluation of drug-drug interactions with erlotinib and gefitinib use in the Military Health System. Fed Pract. 2023;40(Suppl 3):S24-S34. doi:10.12788/fp.0401
- Luong TT, Shou KJ, Reinhard BJ, Kigelman OF, Greenfield KM. Paclitaxel drug-drug interactions in the Military Health System. Fed Pract. 2024;41(Suppl 3):S70-S82. doi:10.12788/fp.0499
- Luong TT, Powers CN, Reinhardt BJ, Weina PJ. Preclinical drug-drug interactions (DDIs) of gefitinib with/without losartan and selective serotonin reuptake inhibitors (SSRIs): citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, and venlafaxine. Curr Res Pharmacol Drug Discov. 2022;3:100112. doi:10.1016/j.crphar.2022.100112
- Luong TT, McAnulty MJ, Evers DL, Reinhardt BJ, Weina PJ. Pre-clinical drug-drug interaction (DDI) of gefitinib or erlotinib with cytochrome P450 (CYP) inhibiting drugs, fluoxetine and/or losartan. Curr Res Toxicol. 2021;2:217- 224. doi:10.1016/j.crtox.2021.05.006,
- Adamo M, Dickie L, Ruhl J. SEER program coding and staging manual 2016. National Cancer Institute; 2016. Accessed April 4, 2025. https://seer.cancer.gov/archive/manuals/2016/SPCSM_2016_maindoc.pdf
- World Health Organization. International classification of diseases for oncology (ICD-O). 3rd ed, 1st revision. World Health Organization; 2013. Accessed April 4, 2025. https://apps.who.int/iris/handle/10665/96612
- Simon S. FDA approves Jelmyto (mitomycin gel) for urothelial cancer. American Cancer Society. April 20, 2020. Accessed April 4, 2025. https://www.cancer.org/cancer/latest-news/fda-approves-jelmyto-mitomycin-gel-for-urothelial-cancer.html
- Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing drugs in oncology (ReDO)- diclofenac as an anti-cancer agent. Ecancermedicalscience. 2016;10:610. doi:10.3332/ecancer.2016.610
- Choi S, Kim S, Park J, Lee SE, Kim C, Kang D. Diclofenac: a nonsteroidal anti-inflammatory drug inducing cancer cell death by inhibiting microtubule polymerization and autophagy flux. Antioxidants (Basel). 2022;11(5):1009. doi:10.3390/antiox11051009
- Tontonoz M. The ABCs of BCG: oldest approved immunotherapy gets new explanation. Memorial Sloan Kettering Cancer Center. July 17, 2020. Accessed April 4, 2025. https://www.mskcc.org/news/oldest-approved-immunotherapy-gets-new-explanation
- Jordan VC. Tamoxifen as the first targeted long-term adjuvant therapy for breast cancer. Endocr Relat Cancer. 2014;21(3):R235-R246. doi:10.1530/ERC-14-0092
- Cole MP, Jones CT, Todd ID. A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474. Br J Cancer. 1971;25(2):270-275. doi:10.1038/bjc.1971.33
- Jordan VC. Tamoxifen: a most unlikely pioneering medicine. Nat Rev Drug Discov. 2003;2(3):205-213. doi:10.1038/nrd1031
- Maximov PY, McDaniel RE, Jordan VC. Tamoxifen: Pioneering Medicine in Breast Cancer. Springer Basel; 2013. Accessed April 4, 2025. https://link.springer.com/book/10.1007/978-3-0348-0664-0
- Taxol (paclitaxel). Prescribing information. Bristol-Myers Squibb Company; 2011. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020262s049lbl.pdf
- Abraxane (paclitaxel). Prescribing information. Celgene Corporation; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021660s047lbl.pdf
- Tarceva (erlotinib). Prescribing information. OSI Pharmaceuticals, LLC; 2016. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021743s025lbl.pdf
- Docetaxel. Prescribing information. Sichuan Hyiyu Pharmaceutical Co.; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215813s000lbl.pdf
- Alimta (pemetrexed). Prescribing information. Teva Pharmaceuticals; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/208419s004lbl.pdf
- Tagrisso (osimertinib). Prescribing information. Astra- Zeneca Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208065s021lbl.pdf
- Iressa (gefitinib). Prescribing information. AstraZeneca Pharmaceuticals; 2018. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/206995s003lbl.pdf
- Kadcyla (ado-trastuzumab emtansine). Prescribing information. Genentech, Inc.; 2013. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125427lbl.pdf
- Alecensa (alectinib). Prescribing information. Genetech, Inc.; 2017. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2017/208434s003lbl.pdf
- Xalkori (crizotinib). Prescribing information. Pfizer Laboratories; 2022. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2022/202570s033lbl.pdf
- Lorbrena (lorlatinib). Prescribing information. Pfizer Laboratories; 2018. Accessed April 14, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210868s000lbl.pdf
- Alunbrig (brigatinib). Prescribing information. Takeda Pharmaceutical Company; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208772s008lbl.pdf
- Rozlytrek (entrectinib). Prescribing information. Genentech, Inc.; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212725s000lbl.pdf
- Herceptin (trastuzumab). Prescribing information. Genentech, Inc.; 2010. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/103792s5250lbl.pdf
- Cybalta (duloxetine). Prescribing information. Eli Lilly and Company; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021427s049lbl.pdf
- Effexor XR (venlafaxine). Prescribing information. Pfizer Wyeth Pharmaceuticals Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020699s112lbl.pdf
- Desyrel (trazodone hydrochloride). Prescribing information. Pragma Pharmaceuticals; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/018207s032lbl.pdf
- Sertraline hydrochloride. Prescribing information. Almatica Pharma LLC; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/215133s000lbl.pdf
- Remeron (mirtazapine). Prescribing information. Merck & Co. Inc; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/020415s029,%20021208s019lbl.pdf
- Celexa (citalopram). Prescribing information. Allergan USA Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020822s041lbl.pdf
- information. GlaxoSmithKline; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020358s066lbl.pdf
- Amitriptyline hydrochloride tablet. Prescribing information. Quality Care Products LLC; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0f12f50f-7087-46e7-a2e6-356b4c566c9f/0f12f50f-7087-46e7-a2e6-356b4c566c9f.xml
- Lexapro (escitalopram). Prescribing information. AbbVie Inc; 2023. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/021323s055,021365s039lbl.pdf
- Fluoxetine. Prescribing information. Edgemont Pharmaceutical, LLC; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/202133s004s005lbl.pdf
- Paxil (paroxetine). Prescribing Information. Apotex Inc; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/020031s077lbl.pdf
- Pamelor (nortriptyline HCl). Prescribing information. Mallinckrodt, Inc; 2012. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2012/018012s029,018013s061lbl.pdf
- Silenor (doxepin). Prescribing information. Currax Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022036s006lbl.pdf
- Tofranil-PM (imipramine pamote). Prescribing information. Mallinckrodt, Inc; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/017090s078lbl.pdf
- Norpramin (desipramine hydrochloride). Prescribing information. Sanofi-aventis U.S. LLC; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/014399s069lbl.pdf
- Khedezla (desvenlafaxine). Prescribing information. Osmotical Pharmaceutical US LLC; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/204683s006lbl.pdf
- Nefazodone hydrochloride. Prescribing information. Bryant Ranch Prepack; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0bd4c34a-4f43-4c84-8b98-1d074cba97d5/0bd4c34a-4f43-4c84-8b98-1d074cba97d5.xml
- Grassi L, Nanni MG, Rodin G, Li M, Caruso R. The use of antidepressants in oncology: a review and practical tips for oncologists. Ann Oncol. 2018;29(1):101-111. doi:10.1093/annonc/mdx526
- Lee E, Park Y, Li D, Rodriguez-Fuguet A, Wang X, Zhang WC. Antidepressant use and lung cancer risk and survival: a meta-analysis of observational studies. Cancer Res Commun. 2023;3(6):1013-1025. doi:10.1158/2767-9764.CRC-23-0003
- Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Arch Gen Psychiatry. 2009;66(8):848 -856. doi:10.1001/archgenpsychiatry.2009.81
- Grattan A, Sullivan MD, Saunders KW, Campbell CI, Von Korff MR. Depression and prescription opioid misuse among chronic opioid therapy recipients with no history of substance abuse. Ann Fam Med. 2012;10(4):304-311. doi:10.1370/afm.1371
- Cowan DT, Wilson-Barnett J, Griffiths P, Allan LG. A survey of chronic noncancer pain patients prescribed opioid analgesics. Pain Med. 2003;4(4):340-351. doi:10.1111/j.1526-4637.2003.03038.x
- Breckenridge J, Clark JD. Patient characteristics associated with opioid versus nonsteroidal anti-inflammatory drug management of chronic low back pain. J Pain. 2003;4(6):344-350. doi:10.1016/s1526-5900(03)00638-2
- Edlund MJ, Martin BC, Devries A, Fan MY, Braden JB, Sullivan MD. Trends in use of opioids for chronic noncancer pain among individuals with mental health and substance use disorders: the TROUP study. Clin J Pain. 2010;26(1):1-8. doi:10.1097/AJP.0b013e3181b99f35
- Sullivan MD, Edlund MJ, Fan MY, DeVries A, Braden JB, Martin BC. Risks for possible and probable opioid misuse among recipients of chronic opioid therapy in commercial and medicaid insurance plans: the TROUP study. Pain. 2010;150(2):332-339. doi:10.1016/j.pain.2010.05.020
- Dunn KM, Saunders KW, Rutter CM, et al. Opioid prescriptions for chronic pain and overdose: a cohort study. Ann Intern Med. 2010;152(2):85-92. doi:10.7326/0003-4819-152-2-201001190-00006
- Haddad P. Do antidepressants have any potential to cause addiction? J Psychopharmacol. 1999;13(3):300- 307. doi:10.1177/026988119901300321
- Lakeview Health Staff. America’s most abused antidepressants. Lakeview Health. January 24, 2004. Accessed April 4, 2025. https://www.lakeviewhealth.com/blog/us-most-abused-antidepressants/
- Greenhouse Treatment Center Editorial Staff. Addiction to antidepressants: is it possible? America Addiction Centers: Greenhouse Treatment Center. Updated April 23, 2024. Accessed April 4, 2025. https://greenhousetreatment.com/prescription-medication/antidepressants/
- National Cancer Institute. Depression (PDQ)-Health Professional Version. Updated July 25, 2024. Accessed April 4, 2025. https://www.cancer.gov/about-cancer/coping/feelings/depression-hp-pdq
- Krebber AM, Buffart LM, Kleijn G, et al. Prevalence of depression in cancer patients: a meta-analysis of diagnostic interviews and self-report instruments. Psychooncology. 2014;23(2):121-130. doi:10.1002/pon.3409
- Xiang X, An R, Gehlert S. Trends in antidepressant use among U.S. cancer survivors, 1999-2012. Psychiatr Serv. 2015;66(6):564. doi:10.1176/appi.ps.201500007
- Hartung TJ, Brähler E, Faller H, et al. The risk of being depressed is significantly higher in cancer patients than in the general population: Prevalence and severity of depressive symptoms across major cancer types. Eur J Cancer. 2017;72:46-53. doi:10.1016/j.ejca.2016.11.017
- Walker J, Holm Hansen C, Martin P, et al. Prevalence of depression in adults with cancer: a systematic review. Ann Oncol. 2013;24(4):895-900. doi:10.1093/annonc/mds575
- Pitman A, Suleman S, Hyde N, Hodgkiss A. Depression and anxiety in patients with cancer. BMJ. 2018;361:k1415. doi:10.1136/bmj.k1415
- Kisely S, Alotiby MKN, Protani MM, Soole R, Arnautovska U, Siskind D. Breast cancer treatment disparities in patients with severe mental illness: a systematic review and meta-analysis. Psychooncology. 2023;32(5):651-662. doi:10.1002/pon.6120
- Massie MJ. Prevalence of depression in patients with cancer. J Natl Cancer Inst Monogr. 2004;(32):57-71. doi:10.1093/jncimonographs/lgh014
- Rodin G, Katz M, Lloyd N, Green E, Mackay JA, Wong RK. Treatment of depression in cancer patients. Curr Oncol. 2007;14(5):180-188. doi:10.3747/co.2007.146
- Wilson KG, Chochinov HM, Skirko MG, et al. Depression and anxiety disorders in palliative cancer care. J Pain Symptom Manage. 2007;33(2):118-129. doi:10.1016/j.jpainsymman.2006.07.016
- Moradi Y, Dowran B, Sepandi M. The global prevalence of depression, suicide ideation, and attempts in the military forces: a systematic review and meta-analysis of cross sectional studies. BMC Psychiatry. 2021;21(1):510. doi:10.1186/s12888-021-03526-2
- Lu CY, Zhang F, Lakoma MD, et al. Changes in antidepressant use by young people and suicidal behavior after FDA warnings and media coverage: quasi-experimental study. BMJ. 2014;348:g3596. doi:10.1136/bmj.g3596
- Friedman RA. Antidepressants’ black-box warning--10 years later. N Engl J Med. 2014;371(18):1666-1668. doi:10.1056/NEJMp1408480
- Sheffler ZM, Patel P, Abdijadid S. Antidepressants. In: StatPearls. StatPearls Publishing. Updated May 26, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK538182/
- Miller JJ. Antidepressants, Part 1: 100 Years and Counting. Accessed April 4, 2025. Psychiatric Times. https:// www.psychiatrictimes.com/view/antidepressants-part-1-100-years-and-counting
- Hillhouse TM, Porter JH. A brief history of the development of antidepressant drugs: from monoamines to glutamate. Exp Clin Psychopharmacol. 2015;23(1):1-21. doi:10.1037/a0038550
- Mitchell PB, Mitchell MS. The management of depression. Part 2. The place of the new antidepressants. Aust Fam Physician. 1994;23(9):1771-1781.
- Sabri MA, Saber-Ayad MM. MAO Inhibitors. In: Stat- Pearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557395/
- Sub Laban T, Saadabadi A. Monoamine Oxidase Inhibitors (MAOI). In: StatPearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK539848/
- Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract. 2004;10(4):239-248. doi:10.1097/00131746-200407000-00005
- Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03
- Moraczewski J, Awosika AO, Aedma KK. Tricyclic Antidepressants. In: StatPearls. StatPearls Publishing. Updated August 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557791/
- Almasi A, Patel P, Meza CE. Doxepin. In: StatPearls. StatPearls Publishing. Updated February 14, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK542306/
- Thour A, Marwaha R. Amitriptyline. In: StatPearls. Stat- Pearls Publishing. Updated July 18, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK537225/
- Radley DC, Finkelstein SN, Stafford RS. Off-label prescribing among office-based physicians. Arch Intern Med. 2006;166(9):1021-1026. doi:10.1001/archinte.166.9.1021
- Tesfaye S, Sloan G, Petrie J, et al. Comparison of amitriptyline supplemented with pregabalin, pregabalin supplemented with amitriptyline, and duloxetine supplemented with pregabalin for the treatment of diabetic peripheral neuropathic pain (OPTION-DM): a multicentre, double-blind, randomised crossover trial. Lancet. 2022;400(10353):680- 690. doi:10.1016/S0140-6736(22)01472-6
- Farag HM, Yunusa I, Goswami H, Sultan I, Doucette JA, Eguale T. Comparison of amitriptyline and US Food and Drug Administration-approved treatments for fibromyalgia: a systematic review and network metaanalysis. JAMA Netw Open. 2022;5(5):e2212939. doi:10.1001/jamanetworkopen.2022.12939
- Merwar G, Gibbons JR, Hosseini SA, Saadabadi A. Nortriptyline. In: StatPearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482214/
- Fayez R, Gupta V. Imipramine. In: StatPearls. StatPearls Publishing. Updated May 22, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557656/
- Jilani TN, Gibbons JR, Faizy RM, Saadabadi A. Mirtazapine. In: StatPearls. StatPearls Publishing. Updated August 28, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK519059/
- Nutt DJ. Tolerability and safety aspects of mirtazapine. Hum Psychopharmacol. 2002;17 Suppl 1:S37-S41. doi:10.1002/hup.388
- Gandotra K, Chen P, Jaskiw GE, Konicki PE, Strohl KP. Effective treatment of insomnia with mirtazapine attenuates concomitant suicidal ideation. J Clin Sleep Med. 2018;14(5):901-902. doi:10.5664/jcsm.7142
- Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249-264. doi:10.1111/j.1527-3458.2001.tb00198.x
- Wang SM, Han C, Bahk WM, et al. Addressing the side effects of contemporary antidepressant drugs: a comprehensive review. Chonnam Med J. 2018;54(2):101-112. doi:10.4068/cmj.2018.54.2.101
- Huecker MR, Smiley A, Saadabadi A. Bupropion. In: StatPearls. StatPearls Publishing. Updated April 9, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470212/
- Hsieh MT, Tseng PT, Wu YC, et al. Effects of different pharmacologic smoking cessation treatments on body weight changes and success rates in patients with nicotine dependence: a network meta-analysis. Obes Rev. 2019;20(6):895- 905. doi:10.1111/obr.12835
- Hankosky ER, Bush HM, Dwoskin LP, et al. Retrospective analysis of health claims to evaluate pharmacotherapies with potential for repurposing: association of bupropion and stimulant use disorder remission. AMIA Annu Symp Proc. 2018;2018:1292-1299.
- Livingstone-Banks J, Norris E, Hartmann-Boyce J, West R, Jarvis M, Hajek P. Relapse prevention interventions for smoking cessation. Cochrane Database Syst Rev. 2019; 2(2):CD003999. doi:10.1002/14651858.CD003999.pub5
- Clayton AH, Kingsberg SA, Goldstein I. Evaluation and management of hypoactive sexual desire disorder. Sex Med. 2018;6(2):59-74. doi:10.1016/j.esxm.2018.01.004
- Verbeeck W, Bekkering GE, Van den Noortgate W, Kramers C. Bupropion for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev. 2017;10(10):CD009504. doi:10.1002/14651858.CD009504.pub2
- Ng QX. A systematic review of the use of bupropion for attention-deficit/hyperactivity disorder in children and adolescents. J Child Adolesc Psychopharmacol. 2017;27(2):112-116. doi:10.1089/cap.2016.0124
- Fava M, Rush AJ, Thase ME, et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim Care Companion J Clin Psychiatry. 2005;7(3):106-113. doi:10.4088/pcc.v07n0305
- Chu A, Wadhwa R. Selective Serotonin Reuptake Inhibitors. In: StatPearls. StatPearls Publishing. Updated May 1, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK554406/
- Singh HK, Saadabadi A. Sertraline. In: StatPearls. Stat- Pearls Publishing. Updated February 13, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK547689/
- MacQueen G, Born L, Steiner M. The selective serotonin reuptake inhibitor sertraline: its profile and use in psychiatric disorders. CNS Drug Rev. 2001;7(1):1-24. doi:10.1111/j.1527-3458.2001.tb00188.x
- Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatments meta-analysis. Lancet. 2009;373(9665):746-758. doi:10.1016/S0140-6736(09)60046-5
- Nelson JC. The STAR*D study: a four-course meal that leaves us wanting more. Am J Psychiatry. 2006;163(11):1864-1866. doi:10.1176/ajp.2006.163.11.1864
- Sharbaf Shoar N, Fariba KA, Padhy RK. Citalopram. In: StatPearls. StatPearls Publishing. Updated November 7, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482222/
- Landy K, Rosani A, Estevez R. Escitalopram. In: Stat- Pearls. StatPearls Publishing. Updated November 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557734/
- Cavanah LR, Ray P, Goldhirsh JL, Huey LY, Piper BJ. Rise of escitalopram and the fall of citalopram. medRxiv. Preprint published online May 8, 2023. doi:10.1101/2023.05.07.23289632
- Sohel AJ, Shutter MC, Patel P, Molla M. Fluoxetine. In: StatPearls. StatPearls Publishing. Updated July 4, 2022. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK459223/
- Wong DT, Perry KW, Bymaster FP. Case history: the discovery of fluoxetine hydrochloride (Prozac). Nat Rev Drug Discov. 2005;4(9):764-774. doi:10.1038/nrd1821
- Shrestha P, Fariba KA, Abdijadid S. Paroxetine. In: Stat- Pearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK526022/
- Naseeruddin R, Rosani A, Marwaha R. Desvenlafaxine. In: StatPearls. StatPearls Publishing. Updated July 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK534829
- Lieberman DZ, Massey SH. Desvenlafaxine in major depressive disorder: an evidence-based review of its place in therapy. Core Evid. 2010;4:67-82. doi:10.2147/ce.s5998
- Withdrawal assessment report for Ellefore (desvenlafaxine). European Medicine Agency. 2009. Accessed April 4, 2025. https://www.ema.europa.eu/en/documents/withdrawal-report/withdrawal-assessment-report-ellefore_en.pdf
- Dhaliwal JS, Spurling BC, Molla M. Duloxetine. In: Stat- Pearls. Statpearls Publishing. Updated May 29, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK549806/
- Hershman DL, Lacchetti C, Dworkin RH, et al. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2014;32(18):1941-1967. doi:10.1200/JCO.2013.54.0914
- Sommer C, Häuser W, Alten R, et al. Medikamentöse Therapie des Fibromyalgiesyndroms. Systematische Übersicht und Metaanalyse [Drug therapy of fibromyalgia syndrome. Systematic review, meta-analysis and guideline]. Schmerz. 2012;26(3):297-310. doi:10.1007/s00482-012-1172-2
- Bril V, England J, Franklin GM, et al. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology. 2011;76(20):1758-1765. doi:10.1212/WNL.0b013e3182166ebe
- Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2010;17(9):1113-e88. doi:10.1111/j.1468-1331.2010.02999.x
- Singh D, Saadabadi A. Venlafaxine. In: StatPearls. StatePearls Publishing. Updated February 26, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK535363/
- Sertraline and venlafaxine: new indication. Prevention of recurrent depression: no advance. Prescrire Int. 2005;14(75):19-20.
- Bruno A, Morabito P, S p i n a E , M u s c a t ello MRl. The role of levomilnacipran in the management of major depressive disorder: a comprehensive review. Curr Neuro pharmacol. 2016;14(2):191-199. doi:10.2174/1570159x14666151117122458
- Shin JJ, Saadabadi A. Trazodone. In: StatPearls. StatPearls Publishing. Updated February 29, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470560/
- Khouzam HR. A review of trazodone use in psychiatric and medical conditions. Postgrad Med. 2017;129(1):140-148. doi:10.1080/00325481.2017.1249265
- Smales ET, Edwards BA, Deyoung PN, et al. Trazodone effects on obstructive sleep apnea and non-REM arousal threshold. Ann Am Thorac Soc. 2015;12(5):758-764. doi:10.1513/AnnalsATS.201408-399OC
- Eckert DJ, Malhotra A, Wellman A, White DP. Trazodone increases the respiratory arousal threshold in patients with obstructive sleep apnea and a low arousal threshold. Sleep. 2014;37(4):811-819. doi:10.5665/sleep.3596
- Schatzberg AF, Nemeroff CB, eds. The American Psychiat ric Association Publishing Textbook of Psychopharmacology. 4th ed. American Psychiatric Publishing; 2009.
- Ruxton K, Woodman RJ, Mangoni AA. Drugs with anticholinergic effects and cognitive impairment, falls and allcause mortality in older adults: a systematic review and meta-analysis. Br J Clin Pharmacol. 2015;80(2):209-220. doi:10.1111/bcp.12617
- Nefazodone. In: LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. National Institute of Diabetes and Digestive and Kidney Diseases. Updated March 6, 2020. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK548179/
- Drugs of Current Interest: Nefazodone. WHO Pharmaceuticals Newsletter:(1). 2003(1):7. https://web.archive.org/web/20150403165029/http:/apps.who.int/medicinedocs/en/d/Js4944e/3.html
- Choi S. Nefazodone (serzone) withdrawn because of hepatotoxicity. CMAJ. 2003;169(11):1187.
- Teva Nefazodone Statement. News release. Teva USA. December 20, 2021. Accessed April 4, 2025. https:// www.tevausa.com/news-and-media/press-releases/teva-nefazodone-statement/
- Levitan MN, Papelbaum M, Nardi AE. Profile of agomelatine and its potential in the treatment of generalized anxiety disorder. Neuropsychiatr Dis Treat. 2015;11:1149- 1155. doi:10.2147/NDT.S67470
- Fu DJ, Ionescu DF, Li X, et al. Esketamine nasal spray for rapid reduction of major depressive disorder symptoms in patients who have active suicidal ideation with intent: double-blind, randomized study (ASPIRE I). J Clin Psychiatry. 2020;81(3):19m13191. doi:10.4088/JCP.19m13191
- Iosifescu DV, Jones A, O’Gorman C, et al. Efficacy and safety of AXS-05 (dextromethorphan-bupropion) in patients with major depressive disorder: a phase 3 randomized clinical trial (GEMINI). J Clin Psychiatry. 2022;83(4): 21m14345. doi:10.4088/JCP.21m14345
- Luong TT, Powers CN, Reinhardt BJ, et al. Retrospective evaluation of drug-drug interactions with erlotinib and gefitinib use in the Military Health System. Fed Pract. 2023;40(Suppl 3):S24-S34. doi:10.12788/fp.0401
- Luong TT, Shou KJ, Reinhard BJ, Kigelman OF, Greenfield KM. Paclitaxel drug-drug interactions in the Military Health System. Fed Pract. 2024;41(Suppl 3):S70-S82. doi:10.12788/fp.0499
- Luong TT, Powers CN, Reinhardt BJ, Weina PJ. Preclinical drug-drug interactions (DDIs) of gefitinib with/without losartan and selective serotonin reuptake inhibitors (SSRIs): citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, and venlafaxine. Curr Res Pharmacol Drug Discov. 2022;3:100112. doi:10.1016/j.crphar.2022.100112
- Luong TT, McAnulty MJ, Evers DL, Reinhardt BJ, Weina PJ. Pre-clinical drug-drug interaction (DDI) of gefitinib or erlotinib with cytochrome P450 (CYP) inhibiting drugs, fluoxetine and/or losartan. Curr Res Toxicol. 2021;2:217- 224. doi:10.1016/j.crtox.2021.05.006,
- Adamo M, Dickie L, Ruhl J. SEER program coding and staging manual 2016. National Cancer Institute; 2016. Accessed April 4, 2025. https://seer.cancer.gov/archive/manuals/2016/SPCSM_2016_maindoc.pdf
- World Health Organization. International classification of diseases for oncology (ICD-O). 3rd ed, 1st revision. World Health Organization; 2013. Accessed April 4, 2025. https://apps.who.int/iris/handle/10665/96612
- Simon S. FDA approves Jelmyto (mitomycin gel) for urothelial cancer. American Cancer Society. April 20, 2020. Accessed April 4, 2025. https://www.cancer.org/cancer/latest-news/fda-approves-jelmyto-mitomycin-gel-for-urothelial-cancer.html
- Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing drugs in oncology (ReDO)- diclofenac as an anti-cancer agent. Ecancermedicalscience. 2016;10:610. doi:10.3332/ecancer.2016.610
- Choi S, Kim S, Park J, Lee SE, Kim C, Kang D. Diclofenac: a nonsteroidal anti-inflammatory drug inducing cancer cell death by inhibiting microtubule polymerization and autophagy flux. Antioxidants (Basel). 2022;11(5):1009. doi:10.3390/antiox11051009
- Tontonoz M. The ABCs of BCG: oldest approved immunotherapy gets new explanation. Memorial Sloan Kettering Cancer Center. July 17, 2020. Accessed April 4, 2025. https://www.mskcc.org/news/oldest-approved-immunotherapy-gets-new-explanation
- Jordan VC. Tamoxifen as the first targeted long-term adjuvant therapy for breast cancer. Endocr Relat Cancer. 2014;21(3):R235-R246. doi:10.1530/ERC-14-0092
- Cole MP, Jones CT, Todd ID. A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474. Br J Cancer. 1971;25(2):270-275. doi:10.1038/bjc.1971.33
- Jordan VC. Tamoxifen: a most unlikely pioneering medicine. Nat Rev Drug Discov. 2003;2(3):205-213. doi:10.1038/nrd1031
- Maximov PY, McDaniel RE, Jordan VC. Tamoxifen: Pioneering Medicine in Breast Cancer. Springer Basel; 2013. Accessed April 4, 2025. https://link.springer.com/book/10.1007/978-3-0348-0664-0
- Taxol (paclitaxel). Prescribing information. Bristol-Myers Squibb Company; 2011. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020262s049lbl.pdf
- Abraxane (paclitaxel). Prescribing information. Celgene Corporation; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021660s047lbl.pdf
- Tarceva (erlotinib). Prescribing information. OSI Pharmaceuticals, LLC; 2016. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021743s025lbl.pdf
- Docetaxel. Prescribing information. Sichuan Hyiyu Pharmaceutical Co.; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215813s000lbl.pdf
- Alimta (pemetrexed). Prescribing information. Teva Pharmaceuticals; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/208419s004lbl.pdf
- Tagrisso (osimertinib). Prescribing information. Astra- Zeneca Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208065s021lbl.pdf
- Iressa (gefitinib). Prescribing information. AstraZeneca Pharmaceuticals; 2018. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/206995s003lbl.pdf
- Kadcyla (ado-trastuzumab emtansine). Prescribing information. Genentech, Inc.; 2013. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125427lbl.pdf
- Alecensa (alectinib). Prescribing information. Genetech, Inc.; 2017. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2017/208434s003lbl.pdf
- Xalkori (crizotinib). Prescribing information. Pfizer Laboratories; 2022. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2022/202570s033lbl.pdf
- Lorbrena (lorlatinib). Prescribing information. Pfizer Laboratories; 2018. Accessed April 14, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210868s000lbl.pdf
- Alunbrig (brigatinib). Prescribing information. Takeda Pharmaceutical Company; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208772s008lbl.pdf
- Rozlytrek (entrectinib). Prescribing information. Genentech, Inc.; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212725s000lbl.pdf
- Herceptin (trastuzumab). Prescribing information. Genentech, Inc.; 2010. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/103792s5250lbl.pdf
- Cybalta (duloxetine). Prescribing information. Eli Lilly and Company; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021427s049lbl.pdf
- Effexor XR (venlafaxine). Prescribing information. Pfizer Wyeth Pharmaceuticals Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020699s112lbl.pdf
- Desyrel (trazodone hydrochloride). Prescribing information. Pragma Pharmaceuticals; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/018207s032lbl.pdf
- Sertraline hydrochloride. Prescribing information. Almatica Pharma LLC; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/215133s000lbl.pdf
- Remeron (mirtazapine). Prescribing information. Merck & Co. Inc; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/020415s029,%20021208s019lbl.pdf
- Celexa (citalopram). Prescribing information. Allergan USA Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020822s041lbl.pdf
- information. GlaxoSmithKline; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020358s066lbl.pdf
- Amitriptyline hydrochloride tablet. Prescribing information. Quality Care Products LLC; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0f12f50f-7087-46e7-a2e6-356b4c566c9f/0f12f50f-7087-46e7-a2e6-356b4c566c9f.xml
- Lexapro (escitalopram). Prescribing information. AbbVie Inc; 2023. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/021323s055,021365s039lbl.pdf
- Fluoxetine. Prescribing information. Edgemont Pharmaceutical, LLC; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/202133s004s005lbl.pdf
- Paxil (paroxetine). Prescribing Information. Apotex Inc; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/020031s077lbl.pdf
- Pamelor (nortriptyline HCl). Prescribing information. Mallinckrodt, Inc; 2012. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2012/018012s029,018013s061lbl.pdf
- Silenor (doxepin). Prescribing information. Currax Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022036s006lbl.pdf
- Tofranil-PM (imipramine pamote). Prescribing information. Mallinckrodt, Inc; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/017090s078lbl.pdf
- Norpramin (desipramine hydrochloride). Prescribing information. Sanofi-aventis U.S. LLC; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/014399s069lbl.pdf
- Khedezla (desvenlafaxine). Prescribing information. Osmotical Pharmaceutical US LLC; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/204683s006lbl.pdf
- Nefazodone hydrochloride. Prescribing information. Bryant Ranch Prepack; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0bd4c34a-4f43-4c84-8b98-1d074cba97d5/0bd4c34a-4f43-4c84-8b98-1d074cba97d5.xml
- Grassi L, Nanni MG, Rodin G, Li M, Caruso R. The use of antidepressants in oncology: a review and practical tips for oncologists. Ann Oncol. 2018;29(1):101-111. doi:10.1093/annonc/mdx526
- Lee E, Park Y, Li D, Rodriguez-Fuguet A, Wang X, Zhang WC. Antidepressant use and lung cancer risk and survival: a meta-analysis of observational studies. Cancer Res Commun. 2023;3(6):1013-1025. doi:10.1158/2767-9764.CRC-23-0003
- Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Arch Gen Psychiatry. 2009;66(8):848 -856. doi:10.1001/archgenpsychiatry.2009.81
- Grattan A, Sullivan MD, Saunders KW, Campbell CI, Von Korff MR. Depression and prescription opioid misuse among chronic opioid therapy recipients with no history of substance abuse. Ann Fam Med. 2012;10(4):304-311. doi:10.1370/afm.1371
- Cowan DT, Wilson-Barnett J, Griffiths P, Allan LG. A survey of chronic noncancer pain patients prescribed opioid analgesics. Pain Med. 2003;4(4):340-351. doi:10.1111/j.1526-4637.2003.03038.x
- Breckenridge J, Clark JD. Patient characteristics associated with opioid versus nonsteroidal anti-inflammatory drug management of chronic low back pain. J Pain. 2003;4(6):344-350. doi:10.1016/s1526-5900(03)00638-2
- Edlund MJ, Martin BC, Devries A, Fan MY, Braden JB, Sullivan MD. Trends in use of opioids for chronic noncancer pain among individuals with mental health and substance use disorders: the TROUP study. Clin J Pain. 2010;26(1):1-8. doi:10.1097/AJP.0b013e3181b99f35
- Sullivan MD, Edlund MJ, Fan MY, DeVries A, Braden JB, Martin BC. Risks for possible and probable opioid misuse among recipients of chronic opioid therapy in commercial and medicaid insurance plans: the TROUP study. Pain. 2010;150(2):332-339. doi:10.1016/j.pain.2010.05.020
- Dunn KM, Saunders KW, Rutter CM, et al. Opioid prescriptions for chronic pain and overdose: a cohort study. Ann Intern Med. 2010;152(2):85-92. doi:10.7326/0003-4819-152-2-201001190-00006
- Haddad P. Do antidepressants have any potential to cause addiction? J Psychopharmacol. 1999;13(3):300- 307. doi:10.1177/026988119901300321
- Lakeview Health Staff. America’s most abused antidepressants. Lakeview Health. January 24, 2004. Accessed April 4, 2025. https://www.lakeviewhealth.com/blog/us-most-abused-antidepressants/
- Greenhouse Treatment Center Editorial Staff. Addiction to antidepressants: is it possible? America Addiction Centers: Greenhouse Treatment Center. Updated April 23, 2024. Accessed April 4, 2025. https://greenhousetreatment.com/prescription-medication/antidepressants/
Assessing the Impact of Antidepressants on Cancer Treatment: A Retrospective Analysis of 14 Antineoplastic Agents
Assessing the Impact of Antidepressants on Cancer Treatment: A Retrospective Analysis of 14 Antineoplastic Agents
Safety and Efficacy of Ezetimibe in Patients With and Without Chronic Kidney Disease at a Pharmacist-Managed Clinic
Statins are widely used to reduce low-density lipoprotein (LDL) and non-high-density lipoprotein (HDL) levels for the prevention of atherosclerotic cardiovascular disease (ASCVD).1 However, despite maximally tolerated statin therapy, many patients may not reach their LDL and non-HDL goals. Some patients may experience adverse events (AEs), particularly muscle-related AEs, which can limit the use of these medications.
The 2022 American College of Cardiology (ACC) expert consensus pathway recommends a goal LDL of < 55 mg/dL in very high-risk patients, defined as those with a history of multiple major ASCVD events or 1 major ASCVD event and multiple high-risk conditions.2 Major ASCVD events include acute coronary syndrome within 12 months, history of myocardial infarction (MI) or ischemic stroke, and symptomatic peripheral arterial disease (ie, claudication with ankle-brachial index < 0.85 or previous revascularization or amputation). Factors for being considered high risk include age > 65 years, heterozygous familial hypercholesterolemia, history of prior coronary artery bypass surgery or percutaneous coronary intervention outside the major ASCVD events, diabetes, hypertension, chronic kidney disease (CKD) (estimated glomerular filtration rate [eGFR] 15-59 mL/min/1.73 m2), current smoking, persistently elevated LDL cholesterol (LDL-C) levels despite maximally tolerated statin therapy and ezetimibe, and history of congestive heart failure.2 For these patients, statin therapy alone may not achieve LDL goal.
The ACC recommends ezetimibe as the initial nonstatin therapy in patients who are not at their goal LDL.2 Ezetimibe works by inhibiting Niemann-Pick C1-Like 1 protein, which causes reduced cholesterol absorption in the small intestine.2,3 Previous studies have shown the benefit of ezetimibe for LDL reduction and ASCVD prevention.4-7 The 2015 IMPROVE-IT study found the combination of simvastatin and ezetimibe resulted in a significantly lower risk of cardiovascular events than simvastatin monotherapy. IMPROVE-IT also reported a further clinical benefit when lower LDL targets (ie, < 55 mg/dL) are achieved, which aligns with the expert consensus pathway recommendations for a lower LDL goal for very high-risk patients.2,5
The RACING trial found that treatment with a moderate-intensity statin and ezetimibe was noninferior to treatment with a high-intensity statin for the primary outcome of occurrence of cardiovascular death, major cardiovascular events, or nonfatal stroke within 3 years. The combination of moderate-intensity statin and ezetimibe achieved lower LDL-C levels and lower incidence of drug intolerance compared to high intensity statin monotherapy.6 The SHARP-CKD study assessed major atherosclerotic events in patients with CKD who had no history of MI or coronary revascularization. The study found that lowering LDL-C with the combination of simvastatin plus ezetimibe safely reduces the risk of major atherosclerotic events in a wide range of patients with CKD.7
Lastly, the 2019 EWTOPIA 75 study found that ezetimibe noted a statistically significant reduction in the incidence of the composite of sudden cardiac death, MI, coronary revascularization, or stroke compared to placebo. Ezetimibe showed benefits in preventing ASCVD events independently of statin therapy.8 These clinical trials provided evidence for the efficacy of ezetimibe for secondary or primary prevention of ASCVD, patients with CKD, and patients who are not at their LDL goal despite maximally tolerated statin therapy.
Reductions in LDL levels with ezetimibe are reported to be 15% to 19% for monotherapy and 13% to 25% when used in combination with a statin.4 Given that the ACC now recommends lower LDL goals, patients may need additional lowering despite taking maximally tolerated statin therapy.2 Additionally, the package insert for ezetimibe reports increased area under the curve (AUC) values of ezetimibe and its metabolites in patients with severe renal disease. It is anticipated that ezetimibe may show an increased reduction of LDL and non-HDL, but there may also be an increased risk for muscle-related AEs.3
This quality-assurance quality improvement project investigated the use of ezetimibe in patients with CKD to determine whether there is further LDL and non-HDL reduction in this patient population. It sought to determine the LDL and non-HDL percentage reduction in patients with and without CKD at the Wilkes-Barre Veterans Affairs Medical Center (WBVAMC) and whether there is an increased risk for muscle-related AEs. Determining the percentage reduction of LDL and non-HDL within this population can help increase use of ezetimibe in patients not at their LDL or non-HDL goal or for those patients unable to tolerate statin therapy.
Methods
This single-center retrospective chart review investigated patients prescribed ezetimibe by a patient aligned care team (PACT) pharmacist at WBVAMC between September 1, 2021, and September 1, 2023. This project was determined to be nonresearch by the Veterans Integrated Service Network 4 multisite institutional review board. Patients were excluded from the review if they started taking ezetimibe outside of the prespecified time frame, if ezetimibe was initiated by a non-WBVAMC PACT pharmacist, or if there was no follow-up lipid panel obtained within 6 months of initiation of ezetimibe.
The primary outcomes were to determine the percentage mean change in LDL and non-HDL reduction and the incidence of muscle-related AEs after initiation of ezetimibe in patients without CKD. The secondary outcomes were to determine the percentage mean change in LDL and non-HDL levels and the incidence of muscle-related AEs after initiation of ezetimibe in patients with CKD. For this study, CKD was defined as a patient having an eGFR 15 to 60 ml/min/1.73 m2. Non-HDL is the combination of LDL-C and very LDL-C and represents all potentially atherogenic particles. The 2022 Expert Consensus Pathway included non-HDL goals in addition to LDL goals.2 Non-HDL cholesterol levels can be used for patients with elevated triglycerides where LDL levels may not be as accurate. To account for instances of elevated triglycerides, this study assessed changes in both LDL and non-HDL levels.
Data were collected from the US Department of Veterans Affairs (VA) Computerized Patient Record System (CPRS) and recorded in a spreadsheet. Collected data included age, sex, race, concomitant cholesterol-lowering medications (statin, proprotein convertase subtilisin/kexin type 9 [PCSK9] inhibitor, bempedoic acid, fish oil, niacin, bile acid sequestrants, and fibrates), baseline lipid panel, lipid panel within 6 months of ezetimibe initiation, and eGFR level. If the patient’s LDL or non-HDL levels worsened on the follow-up lipid panel, their baseline LDL and non-HDL levels were used to calculate the percentage reduction; thus, the percentage reduction would be 0%. This strategy was used in prior research, notably the IMPROVE-IT and SHARP-CKD trials.
Ezetimibe 5 mg once daily was used in this study based on a 2008 VA study that evaluated the use of ezetimibe 5 mg vs ezetimibe 10 mg and the percentage reduction of LDL with each dose. The study found no significant difference between the 5 mg and 10 mg dose.9 Most patients included in this study received the 5 mg dose.
Results
This retrospective chart review consisted of 173 patients, 137 (79.2%) without CKD and 36 (20.8%) with CKD at baseline. The mean age was 69.6 years, 155 (89.6%) patients were male, and 18 (10.4%) were female. There were 164 concomitant medications, including 115 patients prescribed a statin and 38 patients prescribed fish oil (Table 1).
Patients without CKD had mean reductions in LDL levels of 23.5% and non-HDL levels of 21.7% (Figure). Patients who had an increase in LDL and non-HDL levels were excluded to control for potential confounding factors such as dietary changes, discontinuation of ezetimibe therapy, nonadherence to ezetimibe, and medication changes that impacted follow-up laboratory tests such as discontinuation of a statin. Fifteen patients experienced an increase in LDL or non-HDL levels. After excluding these patients, those without CKD had a mean reduction in LDL levels of 28.0% and non-HDL levels of 25.5%. Nineteen (13.9%) patients without CKD experienced a muscle-related AE (Table 2). One patient discontinued ezetimibe and statin use following a Lyme disease diagnosis due to concerns over potential muscle-related AEs.
Patients with CKD had a mean reduction in LDL and non-HDL levels of 27.0% and 24.8%, respectively. Patients with an increase in LDL or non-HDL levels were also excluded to help control for potential confounding factors. After excluding 4 patients with increased LDL and non-HDL levels, the mean reduction in LDL and non-HDL levels was 30.5% and 27.5%, respectively. Five (13.9%) patients with CKD experienced muscle-related AEs thought to be due to ezetimibe. Other AEs (eg, urticaria, diarrhea, reflux, dizziness, headache, upset stomach) were reported that led to discontinuation of ezetimibe, but only muscle-related AEs were analyzed.
Discussion
This retrospective chart review found larger reductions in LDL and non-HDL levels for patients with CKD than reported in the literature.4 Based on the findings that indicate a greater cholesterol reduction with ezetimibe, the results suggest an underutilization of ezetimibe in clinical practice, which may be due to clinicians favoring statin therapy and overlooking ezetimibe as a viable option based on recommendation in earlier guidelines. The 2022 guidelines transitioned from a statin focus to a focus on LDL targets and goals.2
According to the ACC, there is evidence to support a direct relationship between LDL-C levels, atherosclerosis progression, and ASCVD event risk.2 Absolute LDL-C level reduction is directly associated with ASCVD risk reduction which supports the LDL hypothesis. There appears to be no specific LDL-C level below which benefit ceases.2 This suggests that lower LDL-C targets (< 55 mg/dL) should be used when clinically indicated. Many patients are either unable to reach their goal LDL levels with statin monotherapy or are unable to tolerate statin therapy at higher doses, which may require additional pharmacotherapy to reach goal LDL-C. The ACC expert consensus pathway recommends ezetimibe as the initial add-on treatment to statins.2 The RACING trial showed the benefit of adding ezetimibe to a moderate-intensity statin vs increasing to a high-intensity statin dose. This trial found patients had lower LDL levels and lower rates of intolerances, which further supports ezetimibe use.6
This quality improvement project assessed LDL and non-HDL level reduction in patients with CKD. As anticipated, there was greater reduction in LDL and non-HDL levels seen in patients with CKD. The SHARP-CKD trial also found reductions in LDL levels with ezetimibe in patients with CKD.7 Given the reduction in LDL and non-HDL levels with ezetimibe in patients with or without CKD, add-on therapy of ezetimibe should be recommended for patients who do not achieve their LDL goals with statin therapy or for patients who intolerant to statin therapy.
The ezetimibe package insert reports myalgias incidence to be < 5% in patients and research has shown up to a 20% incidence of muscle-related AEs with statin therapy.3,10 Based on the package information reporting increased AUC values of ezetimibe and its metabolites in patients with severe renal disease, it was anticipated there may be an increased risk of muscle-related AEs in patients with CKD.3 However, this study found the same incidence of muscle-related AEs in patients with and without CKD. Previous research on statin-intolerant patients found the incidence of muscle-related AEs with ezetimibe to be 23.0% and 28.8%.11,12 This increased incidence of muscle-related AEs may be the result of including patients with a history of statin intolerance. Collectively, data from clinical trials and this study indicate that patients with prior intolerances to statins appear to have a higher likelihood of developing a muscle-related AEs with ezetimibe.11,12 Clinicians and patients should be educated on the potential for these AEs and be aware that the likelihood may be greater if there is a history of statin intolerance. To our knowledge, this was the first study to evaluate muscle-related AEs with ezetimibe in patients with and without CKD.
Limitations
This retrospective chart review was performed over a prespecified period and only patients initiated on ezetimibe by a PACT pharmacist were included. This study did not assess the percentage of LDL reduction in patients on concomitant statins vs those who were not on concomitant statins. The study only included 173 patients. Additionally, the study was primarily composed of White men and may not be representative of other populations. In addition, veterans may not be representative of the general population given their high comorbidity burden and other exposures. Some reported muscle-related AEs associated with ezetimibe may be attributed to the nocebo effect.
Conclusions
The results of this retrospective chart review suggest there may be a larger mean reduction in LDL and non-HDL levels seen with ezetimibe therapy than reported within the literature. There was a larger mean reduction in LDL and non-HDL levels in patients with CKD than in patients without CKD. Additionally, there were the same rates of muscle-related AEs with ezetimibe therapy in patients with and without CKD. The rates of muscle-related AEs with ezetimibe therapy were higher than reported in the medication’s package insert, but lower than reported in literature that included statin-intolerant patients. These results indicate there may be a benefit to an increase in use of ezetimibe in clinical practice due to its increased effectiveness and safety in patients with and without CKD. Ultimately, this can help patients achieve their LDL goals as recommended by ACC clinical practice guidelines.
Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2019;73(24) e285-e350. doi:10.1016/j.jacc.2018.11.003
Writing Committee, Lloyd-Jones DM, Morris PB, et al. 2022 ACC expert consensus decision pathway on the role of nonstatin therapies for LDL-cholesterol lowering in the management of atherosclerotic cardiovascular disease risk: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2022;80(14):1366-1418. doi:10.1016/j.jacc.2022.07.006
US Food and Drug Administration. Ezetimibe. 2007. Accessed April 1, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/021445s019lbl.pdf
Singh A, Cho LS. Nonstatin therapy to reduce low-density lipoprotein cholesterol and improve cardiovascular outcomes. Cleve Clin J Med. 2024;91(1):53-63. doi:10.3949/ccjm.91a.23058
Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372(25):2387-2397. doi:10.1056/NEJMoa1410489
Kim B, Hong S, Lee Y, et al. Long-term efficacy and safety of moderate-intensity statin with ezetimibe combination therapy versus high-intensity statin monotherapy in patients with atherosclerotic cardiovascular disease (RACING): a randomised, open-label, non-inferiority trial. Lancet. 2022;400(10349):380-390. doi:10.1016/S0140-6736(22)00916-3
Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet. 2011;377(9784):2181-2192. doi:10.1016/S0140-6736(11)60739-3
Ouchi Y, Sasaki J, Arai H, et al. Ezetimibe lipid-lowering trial on prevention of atherosclerotic cardiovascular disease in 75 or older (EWTOPIA 75): a randomized, controlled trial. Circulation. 2019;140:992-1003. doi:10.1161/CIRCULATIONAHA.118.039415
Baruch L, Gupta B, Lieberman-Blum SS, Agarwal S, Eng C. Ezetimibe 5 and 10 mg for lowering LDL-C: potential billion-dollar savings with improved tolerability. Am J Manag Care. 2008;14(10):637-641. https://www.ajmc.com/view/oct08-3644p637-641
Stroes ES, Thompson PD, Corsini A, et al. Statin-associated muscle symptoms: impact on statin therapy-European Atherosclerosis Society Consensus Panel Statement on Assessment, Aetiology and Management. Eur Heart J. 2015;36(17):1012-1022. doi:10.1093/eurheartj/ehv043
Stroes E, Colquhoun D, Sullivan D, et al. Anti-PCSK9 antibody effectively lowers cholesterol in patients with statin intolerance: the GAUSS-2 randomized, placebo-controlled phase 3 clinical trial of evolocumab. J Am Coll Cardiol. 2014;63(23):2541-2548. doi:10.1016/j.jacc.2014.03.019
Nissen SE, Stroes E, Dent-Acosta RE, et al. Efficacy and tolerability of evolocumab vs ezetimibe in patients with muscle-related statin intolerance: the GAUSS-3 randomized clinical trial. JAMA. 2016;315(15):1580-1590. doi:10.1001/jama.2016.3608
Statins are widely used to reduce low-density lipoprotein (LDL) and non-high-density lipoprotein (HDL) levels for the prevention of atherosclerotic cardiovascular disease (ASCVD).1 However, despite maximally tolerated statin therapy, many patients may not reach their LDL and non-HDL goals. Some patients may experience adverse events (AEs), particularly muscle-related AEs, which can limit the use of these medications.
The 2022 American College of Cardiology (ACC) expert consensus pathway recommends a goal LDL of < 55 mg/dL in very high-risk patients, defined as those with a history of multiple major ASCVD events or 1 major ASCVD event and multiple high-risk conditions.2 Major ASCVD events include acute coronary syndrome within 12 months, history of myocardial infarction (MI) or ischemic stroke, and symptomatic peripheral arterial disease (ie, claudication with ankle-brachial index < 0.85 or previous revascularization or amputation). Factors for being considered high risk include age > 65 years, heterozygous familial hypercholesterolemia, history of prior coronary artery bypass surgery or percutaneous coronary intervention outside the major ASCVD events, diabetes, hypertension, chronic kidney disease (CKD) (estimated glomerular filtration rate [eGFR] 15-59 mL/min/1.73 m2), current smoking, persistently elevated LDL cholesterol (LDL-C) levels despite maximally tolerated statin therapy and ezetimibe, and history of congestive heart failure.2 For these patients, statin therapy alone may not achieve LDL goal.
The ACC recommends ezetimibe as the initial nonstatin therapy in patients who are not at their goal LDL.2 Ezetimibe works by inhibiting Niemann-Pick C1-Like 1 protein, which causes reduced cholesterol absorption in the small intestine.2,3 Previous studies have shown the benefit of ezetimibe for LDL reduction and ASCVD prevention.4-7 The 2015 IMPROVE-IT study found the combination of simvastatin and ezetimibe resulted in a significantly lower risk of cardiovascular events than simvastatin monotherapy. IMPROVE-IT also reported a further clinical benefit when lower LDL targets (ie, < 55 mg/dL) are achieved, which aligns with the expert consensus pathway recommendations for a lower LDL goal for very high-risk patients.2,5
The RACING trial found that treatment with a moderate-intensity statin and ezetimibe was noninferior to treatment with a high-intensity statin for the primary outcome of occurrence of cardiovascular death, major cardiovascular events, or nonfatal stroke within 3 years. The combination of moderate-intensity statin and ezetimibe achieved lower LDL-C levels and lower incidence of drug intolerance compared to high intensity statin monotherapy.6 The SHARP-CKD study assessed major atherosclerotic events in patients with CKD who had no history of MI or coronary revascularization. The study found that lowering LDL-C with the combination of simvastatin plus ezetimibe safely reduces the risk of major atherosclerotic events in a wide range of patients with CKD.7
Lastly, the 2019 EWTOPIA 75 study found that ezetimibe noted a statistically significant reduction in the incidence of the composite of sudden cardiac death, MI, coronary revascularization, or stroke compared to placebo. Ezetimibe showed benefits in preventing ASCVD events independently of statin therapy.8 These clinical trials provided evidence for the efficacy of ezetimibe for secondary or primary prevention of ASCVD, patients with CKD, and patients who are not at their LDL goal despite maximally tolerated statin therapy.
Reductions in LDL levels with ezetimibe are reported to be 15% to 19% for monotherapy and 13% to 25% when used in combination with a statin.4 Given that the ACC now recommends lower LDL goals, patients may need additional lowering despite taking maximally tolerated statin therapy.2 Additionally, the package insert for ezetimibe reports increased area under the curve (AUC) values of ezetimibe and its metabolites in patients with severe renal disease. It is anticipated that ezetimibe may show an increased reduction of LDL and non-HDL, but there may also be an increased risk for muscle-related AEs.3
This quality-assurance quality improvement project investigated the use of ezetimibe in patients with CKD to determine whether there is further LDL and non-HDL reduction in this patient population. It sought to determine the LDL and non-HDL percentage reduction in patients with and without CKD at the Wilkes-Barre Veterans Affairs Medical Center (WBVAMC) and whether there is an increased risk for muscle-related AEs. Determining the percentage reduction of LDL and non-HDL within this population can help increase use of ezetimibe in patients not at their LDL or non-HDL goal or for those patients unable to tolerate statin therapy.
Methods
This single-center retrospective chart review investigated patients prescribed ezetimibe by a patient aligned care team (PACT) pharmacist at WBVAMC between September 1, 2021, and September 1, 2023. This project was determined to be nonresearch by the Veterans Integrated Service Network 4 multisite institutional review board. Patients were excluded from the review if they started taking ezetimibe outside of the prespecified time frame, if ezetimibe was initiated by a non-WBVAMC PACT pharmacist, or if there was no follow-up lipid panel obtained within 6 months of initiation of ezetimibe.
The primary outcomes were to determine the percentage mean change in LDL and non-HDL reduction and the incidence of muscle-related AEs after initiation of ezetimibe in patients without CKD. The secondary outcomes were to determine the percentage mean change in LDL and non-HDL levels and the incidence of muscle-related AEs after initiation of ezetimibe in patients with CKD. For this study, CKD was defined as a patient having an eGFR 15 to 60 ml/min/1.73 m2. Non-HDL is the combination of LDL-C and very LDL-C and represents all potentially atherogenic particles. The 2022 Expert Consensus Pathway included non-HDL goals in addition to LDL goals.2 Non-HDL cholesterol levels can be used for patients with elevated triglycerides where LDL levels may not be as accurate. To account for instances of elevated triglycerides, this study assessed changes in both LDL and non-HDL levels.
Data were collected from the US Department of Veterans Affairs (VA) Computerized Patient Record System (CPRS) and recorded in a spreadsheet. Collected data included age, sex, race, concomitant cholesterol-lowering medications (statin, proprotein convertase subtilisin/kexin type 9 [PCSK9] inhibitor, bempedoic acid, fish oil, niacin, bile acid sequestrants, and fibrates), baseline lipid panel, lipid panel within 6 months of ezetimibe initiation, and eGFR level. If the patient’s LDL or non-HDL levels worsened on the follow-up lipid panel, their baseline LDL and non-HDL levels were used to calculate the percentage reduction; thus, the percentage reduction would be 0%. This strategy was used in prior research, notably the IMPROVE-IT and SHARP-CKD trials.
Ezetimibe 5 mg once daily was used in this study based on a 2008 VA study that evaluated the use of ezetimibe 5 mg vs ezetimibe 10 mg and the percentage reduction of LDL with each dose. The study found no significant difference between the 5 mg and 10 mg dose.9 Most patients included in this study received the 5 mg dose.
Results
This retrospective chart review consisted of 173 patients, 137 (79.2%) without CKD and 36 (20.8%) with CKD at baseline. The mean age was 69.6 years, 155 (89.6%) patients were male, and 18 (10.4%) were female. There were 164 concomitant medications, including 115 patients prescribed a statin and 38 patients prescribed fish oil (Table 1).
Patients without CKD had mean reductions in LDL levels of 23.5% and non-HDL levels of 21.7% (Figure). Patients who had an increase in LDL and non-HDL levels were excluded to control for potential confounding factors such as dietary changes, discontinuation of ezetimibe therapy, nonadherence to ezetimibe, and medication changes that impacted follow-up laboratory tests such as discontinuation of a statin. Fifteen patients experienced an increase in LDL or non-HDL levels. After excluding these patients, those without CKD had a mean reduction in LDL levels of 28.0% and non-HDL levels of 25.5%. Nineteen (13.9%) patients without CKD experienced a muscle-related AE (Table 2). One patient discontinued ezetimibe and statin use following a Lyme disease diagnosis due to concerns over potential muscle-related AEs.
Patients with CKD had a mean reduction in LDL and non-HDL levels of 27.0% and 24.8%, respectively. Patients with an increase in LDL or non-HDL levels were also excluded to help control for potential confounding factors. After excluding 4 patients with increased LDL and non-HDL levels, the mean reduction in LDL and non-HDL levels was 30.5% and 27.5%, respectively. Five (13.9%) patients with CKD experienced muscle-related AEs thought to be due to ezetimibe. Other AEs (eg, urticaria, diarrhea, reflux, dizziness, headache, upset stomach) were reported that led to discontinuation of ezetimibe, but only muscle-related AEs were analyzed.
Discussion
This retrospective chart review found larger reductions in LDL and non-HDL levels for patients with CKD than reported in the literature.4 Based on the findings that indicate a greater cholesterol reduction with ezetimibe, the results suggest an underutilization of ezetimibe in clinical practice, which may be due to clinicians favoring statin therapy and overlooking ezetimibe as a viable option based on recommendation in earlier guidelines. The 2022 guidelines transitioned from a statin focus to a focus on LDL targets and goals.2
According to the ACC, there is evidence to support a direct relationship between LDL-C levels, atherosclerosis progression, and ASCVD event risk.2 Absolute LDL-C level reduction is directly associated with ASCVD risk reduction which supports the LDL hypothesis. There appears to be no specific LDL-C level below which benefit ceases.2 This suggests that lower LDL-C targets (< 55 mg/dL) should be used when clinically indicated. Many patients are either unable to reach their goal LDL levels with statin monotherapy or are unable to tolerate statin therapy at higher doses, which may require additional pharmacotherapy to reach goal LDL-C. The ACC expert consensus pathway recommends ezetimibe as the initial add-on treatment to statins.2 The RACING trial showed the benefit of adding ezetimibe to a moderate-intensity statin vs increasing to a high-intensity statin dose. This trial found patients had lower LDL levels and lower rates of intolerances, which further supports ezetimibe use.6
This quality improvement project assessed LDL and non-HDL level reduction in patients with CKD. As anticipated, there was greater reduction in LDL and non-HDL levels seen in patients with CKD. The SHARP-CKD trial also found reductions in LDL levels with ezetimibe in patients with CKD.7 Given the reduction in LDL and non-HDL levels with ezetimibe in patients with or without CKD, add-on therapy of ezetimibe should be recommended for patients who do not achieve their LDL goals with statin therapy or for patients who intolerant to statin therapy.
The ezetimibe package insert reports myalgias incidence to be < 5% in patients and research has shown up to a 20% incidence of muscle-related AEs with statin therapy.3,10 Based on the package information reporting increased AUC values of ezetimibe and its metabolites in patients with severe renal disease, it was anticipated there may be an increased risk of muscle-related AEs in patients with CKD.3 However, this study found the same incidence of muscle-related AEs in patients with and without CKD. Previous research on statin-intolerant patients found the incidence of muscle-related AEs with ezetimibe to be 23.0% and 28.8%.11,12 This increased incidence of muscle-related AEs may be the result of including patients with a history of statin intolerance. Collectively, data from clinical trials and this study indicate that patients with prior intolerances to statins appear to have a higher likelihood of developing a muscle-related AEs with ezetimibe.11,12 Clinicians and patients should be educated on the potential for these AEs and be aware that the likelihood may be greater if there is a history of statin intolerance. To our knowledge, this was the first study to evaluate muscle-related AEs with ezetimibe in patients with and without CKD.
Limitations
This retrospective chart review was performed over a prespecified period and only patients initiated on ezetimibe by a PACT pharmacist were included. This study did not assess the percentage of LDL reduction in patients on concomitant statins vs those who were not on concomitant statins. The study only included 173 patients. Additionally, the study was primarily composed of White men and may not be representative of other populations. In addition, veterans may not be representative of the general population given their high comorbidity burden and other exposures. Some reported muscle-related AEs associated with ezetimibe may be attributed to the nocebo effect.
Conclusions
The results of this retrospective chart review suggest there may be a larger mean reduction in LDL and non-HDL levels seen with ezetimibe therapy than reported within the literature. There was a larger mean reduction in LDL and non-HDL levels in patients with CKD than in patients without CKD. Additionally, there were the same rates of muscle-related AEs with ezetimibe therapy in patients with and without CKD. The rates of muscle-related AEs with ezetimibe therapy were higher than reported in the medication’s package insert, but lower than reported in literature that included statin-intolerant patients. These results indicate there may be a benefit to an increase in use of ezetimibe in clinical practice due to its increased effectiveness and safety in patients with and without CKD. Ultimately, this can help patients achieve their LDL goals as recommended by ACC clinical practice guidelines.
Statins are widely used to reduce low-density lipoprotein (LDL) and non-high-density lipoprotein (HDL) levels for the prevention of atherosclerotic cardiovascular disease (ASCVD).1 However, despite maximally tolerated statin therapy, many patients may not reach their LDL and non-HDL goals. Some patients may experience adverse events (AEs), particularly muscle-related AEs, which can limit the use of these medications.
The 2022 American College of Cardiology (ACC) expert consensus pathway recommends a goal LDL of < 55 mg/dL in very high-risk patients, defined as those with a history of multiple major ASCVD events or 1 major ASCVD event and multiple high-risk conditions.2 Major ASCVD events include acute coronary syndrome within 12 months, history of myocardial infarction (MI) or ischemic stroke, and symptomatic peripheral arterial disease (ie, claudication with ankle-brachial index < 0.85 or previous revascularization or amputation). Factors for being considered high risk include age > 65 years, heterozygous familial hypercholesterolemia, history of prior coronary artery bypass surgery or percutaneous coronary intervention outside the major ASCVD events, diabetes, hypertension, chronic kidney disease (CKD) (estimated glomerular filtration rate [eGFR] 15-59 mL/min/1.73 m2), current smoking, persistently elevated LDL cholesterol (LDL-C) levels despite maximally tolerated statin therapy and ezetimibe, and history of congestive heart failure.2 For these patients, statin therapy alone may not achieve LDL goal.
The ACC recommends ezetimibe as the initial nonstatin therapy in patients who are not at their goal LDL.2 Ezetimibe works by inhibiting Niemann-Pick C1-Like 1 protein, which causes reduced cholesterol absorption in the small intestine.2,3 Previous studies have shown the benefit of ezetimibe for LDL reduction and ASCVD prevention.4-7 The 2015 IMPROVE-IT study found the combination of simvastatin and ezetimibe resulted in a significantly lower risk of cardiovascular events than simvastatin monotherapy. IMPROVE-IT also reported a further clinical benefit when lower LDL targets (ie, < 55 mg/dL) are achieved, which aligns with the expert consensus pathway recommendations for a lower LDL goal for very high-risk patients.2,5
The RACING trial found that treatment with a moderate-intensity statin and ezetimibe was noninferior to treatment with a high-intensity statin for the primary outcome of occurrence of cardiovascular death, major cardiovascular events, or nonfatal stroke within 3 years. The combination of moderate-intensity statin and ezetimibe achieved lower LDL-C levels and lower incidence of drug intolerance compared to high intensity statin monotherapy.6 The SHARP-CKD study assessed major atherosclerotic events in patients with CKD who had no history of MI or coronary revascularization. The study found that lowering LDL-C with the combination of simvastatin plus ezetimibe safely reduces the risk of major atherosclerotic events in a wide range of patients with CKD.7
Lastly, the 2019 EWTOPIA 75 study found that ezetimibe noted a statistically significant reduction in the incidence of the composite of sudden cardiac death, MI, coronary revascularization, or stroke compared to placebo. Ezetimibe showed benefits in preventing ASCVD events independently of statin therapy.8 These clinical trials provided evidence for the efficacy of ezetimibe for secondary or primary prevention of ASCVD, patients with CKD, and patients who are not at their LDL goal despite maximally tolerated statin therapy.
Reductions in LDL levels with ezetimibe are reported to be 15% to 19% for monotherapy and 13% to 25% when used in combination with a statin.4 Given that the ACC now recommends lower LDL goals, patients may need additional lowering despite taking maximally tolerated statin therapy.2 Additionally, the package insert for ezetimibe reports increased area under the curve (AUC) values of ezetimibe and its metabolites in patients with severe renal disease. It is anticipated that ezetimibe may show an increased reduction of LDL and non-HDL, but there may also be an increased risk for muscle-related AEs.3
This quality-assurance quality improvement project investigated the use of ezetimibe in patients with CKD to determine whether there is further LDL and non-HDL reduction in this patient population. It sought to determine the LDL and non-HDL percentage reduction in patients with and without CKD at the Wilkes-Barre Veterans Affairs Medical Center (WBVAMC) and whether there is an increased risk for muscle-related AEs. Determining the percentage reduction of LDL and non-HDL within this population can help increase use of ezetimibe in patients not at their LDL or non-HDL goal or for those patients unable to tolerate statin therapy.
Methods
This single-center retrospective chart review investigated patients prescribed ezetimibe by a patient aligned care team (PACT) pharmacist at WBVAMC between September 1, 2021, and September 1, 2023. This project was determined to be nonresearch by the Veterans Integrated Service Network 4 multisite institutional review board. Patients were excluded from the review if they started taking ezetimibe outside of the prespecified time frame, if ezetimibe was initiated by a non-WBVAMC PACT pharmacist, or if there was no follow-up lipid panel obtained within 6 months of initiation of ezetimibe.
The primary outcomes were to determine the percentage mean change in LDL and non-HDL reduction and the incidence of muscle-related AEs after initiation of ezetimibe in patients without CKD. The secondary outcomes were to determine the percentage mean change in LDL and non-HDL levels and the incidence of muscle-related AEs after initiation of ezetimibe in patients with CKD. For this study, CKD was defined as a patient having an eGFR 15 to 60 ml/min/1.73 m2. Non-HDL is the combination of LDL-C and very LDL-C and represents all potentially atherogenic particles. The 2022 Expert Consensus Pathway included non-HDL goals in addition to LDL goals.2 Non-HDL cholesterol levels can be used for patients with elevated triglycerides where LDL levels may not be as accurate. To account for instances of elevated triglycerides, this study assessed changes in both LDL and non-HDL levels.
Data were collected from the US Department of Veterans Affairs (VA) Computerized Patient Record System (CPRS) and recorded in a spreadsheet. Collected data included age, sex, race, concomitant cholesterol-lowering medications (statin, proprotein convertase subtilisin/kexin type 9 [PCSK9] inhibitor, bempedoic acid, fish oil, niacin, bile acid sequestrants, and fibrates), baseline lipid panel, lipid panel within 6 months of ezetimibe initiation, and eGFR level. If the patient’s LDL or non-HDL levels worsened on the follow-up lipid panel, their baseline LDL and non-HDL levels were used to calculate the percentage reduction; thus, the percentage reduction would be 0%. This strategy was used in prior research, notably the IMPROVE-IT and SHARP-CKD trials.
Ezetimibe 5 mg once daily was used in this study based on a 2008 VA study that evaluated the use of ezetimibe 5 mg vs ezetimibe 10 mg and the percentage reduction of LDL with each dose. The study found no significant difference between the 5 mg and 10 mg dose.9 Most patients included in this study received the 5 mg dose.
Results
This retrospective chart review consisted of 173 patients, 137 (79.2%) without CKD and 36 (20.8%) with CKD at baseline. The mean age was 69.6 years, 155 (89.6%) patients were male, and 18 (10.4%) were female. There were 164 concomitant medications, including 115 patients prescribed a statin and 38 patients prescribed fish oil (Table 1).
Patients without CKD had mean reductions in LDL levels of 23.5% and non-HDL levels of 21.7% (Figure). Patients who had an increase in LDL and non-HDL levels were excluded to control for potential confounding factors such as dietary changes, discontinuation of ezetimibe therapy, nonadherence to ezetimibe, and medication changes that impacted follow-up laboratory tests such as discontinuation of a statin. Fifteen patients experienced an increase in LDL or non-HDL levels. After excluding these patients, those without CKD had a mean reduction in LDL levels of 28.0% and non-HDL levels of 25.5%. Nineteen (13.9%) patients without CKD experienced a muscle-related AE (Table 2). One patient discontinued ezetimibe and statin use following a Lyme disease diagnosis due to concerns over potential muscle-related AEs.
Patients with CKD had a mean reduction in LDL and non-HDL levels of 27.0% and 24.8%, respectively. Patients with an increase in LDL or non-HDL levels were also excluded to help control for potential confounding factors. After excluding 4 patients with increased LDL and non-HDL levels, the mean reduction in LDL and non-HDL levels was 30.5% and 27.5%, respectively. Five (13.9%) patients with CKD experienced muscle-related AEs thought to be due to ezetimibe. Other AEs (eg, urticaria, diarrhea, reflux, dizziness, headache, upset stomach) were reported that led to discontinuation of ezetimibe, but only muscle-related AEs were analyzed.
Discussion
This retrospective chart review found larger reductions in LDL and non-HDL levels for patients with CKD than reported in the literature.4 Based on the findings that indicate a greater cholesterol reduction with ezetimibe, the results suggest an underutilization of ezetimibe in clinical practice, which may be due to clinicians favoring statin therapy and overlooking ezetimibe as a viable option based on recommendation in earlier guidelines. The 2022 guidelines transitioned from a statin focus to a focus on LDL targets and goals.2
According to the ACC, there is evidence to support a direct relationship between LDL-C levels, atherosclerosis progression, and ASCVD event risk.2 Absolute LDL-C level reduction is directly associated with ASCVD risk reduction which supports the LDL hypothesis. There appears to be no specific LDL-C level below which benefit ceases.2 This suggests that lower LDL-C targets (< 55 mg/dL) should be used when clinically indicated. Many patients are either unable to reach their goal LDL levels with statin monotherapy or are unable to tolerate statin therapy at higher doses, which may require additional pharmacotherapy to reach goal LDL-C. The ACC expert consensus pathway recommends ezetimibe as the initial add-on treatment to statins.2 The RACING trial showed the benefit of adding ezetimibe to a moderate-intensity statin vs increasing to a high-intensity statin dose. This trial found patients had lower LDL levels and lower rates of intolerances, which further supports ezetimibe use.6
This quality improvement project assessed LDL and non-HDL level reduction in patients with CKD. As anticipated, there was greater reduction in LDL and non-HDL levels seen in patients with CKD. The SHARP-CKD trial also found reductions in LDL levels with ezetimibe in patients with CKD.7 Given the reduction in LDL and non-HDL levels with ezetimibe in patients with or without CKD, add-on therapy of ezetimibe should be recommended for patients who do not achieve their LDL goals with statin therapy or for patients who intolerant to statin therapy.
The ezetimibe package insert reports myalgias incidence to be < 5% in patients and research has shown up to a 20% incidence of muscle-related AEs with statin therapy.3,10 Based on the package information reporting increased AUC values of ezetimibe and its metabolites in patients with severe renal disease, it was anticipated there may be an increased risk of muscle-related AEs in patients with CKD.3 However, this study found the same incidence of muscle-related AEs in patients with and without CKD. Previous research on statin-intolerant patients found the incidence of muscle-related AEs with ezetimibe to be 23.0% and 28.8%.11,12 This increased incidence of muscle-related AEs may be the result of including patients with a history of statin intolerance. Collectively, data from clinical trials and this study indicate that patients with prior intolerances to statins appear to have a higher likelihood of developing a muscle-related AEs with ezetimibe.11,12 Clinicians and patients should be educated on the potential for these AEs and be aware that the likelihood may be greater if there is a history of statin intolerance. To our knowledge, this was the first study to evaluate muscle-related AEs with ezetimibe in patients with and without CKD.
Limitations
This retrospective chart review was performed over a prespecified period and only patients initiated on ezetimibe by a PACT pharmacist were included. This study did not assess the percentage of LDL reduction in patients on concomitant statins vs those who were not on concomitant statins. The study only included 173 patients. Additionally, the study was primarily composed of White men and may not be representative of other populations. In addition, veterans may not be representative of the general population given their high comorbidity burden and other exposures. Some reported muscle-related AEs associated with ezetimibe may be attributed to the nocebo effect.
Conclusions
The results of this retrospective chart review suggest there may be a larger mean reduction in LDL and non-HDL levels seen with ezetimibe therapy than reported within the literature. There was a larger mean reduction in LDL and non-HDL levels in patients with CKD than in patients without CKD. Additionally, there were the same rates of muscle-related AEs with ezetimibe therapy in patients with and without CKD. The rates of muscle-related AEs with ezetimibe therapy were higher than reported in the medication’s package insert, but lower than reported in literature that included statin-intolerant patients. These results indicate there may be a benefit to an increase in use of ezetimibe in clinical practice due to its increased effectiveness and safety in patients with and without CKD. Ultimately, this can help patients achieve their LDL goals as recommended by ACC clinical practice guidelines.
Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2019;73(24) e285-e350. doi:10.1016/j.jacc.2018.11.003
Writing Committee, Lloyd-Jones DM, Morris PB, et al. 2022 ACC expert consensus decision pathway on the role of nonstatin therapies for LDL-cholesterol lowering in the management of atherosclerotic cardiovascular disease risk: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2022;80(14):1366-1418. doi:10.1016/j.jacc.2022.07.006
US Food and Drug Administration. Ezetimibe. 2007. Accessed April 1, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/021445s019lbl.pdf
Singh A, Cho LS. Nonstatin therapy to reduce low-density lipoprotein cholesterol and improve cardiovascular outcomes. Cleve Clin J Med. 2024;91(1):53-63. doi:10.3949/ccjm.91a.23058
Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372(25):2387-2397. doi:10.1056/NEJMoa1410489
Kim B, Hong S, Lee Y, et al. Long-term efficacy and safety of moderate-intensity statin with ezetimibe combination therapy versus high-intensity statin monotherapy in patients with atherosclerotic cardiovascular disease (RACING): a randomised, open-label, non-inferiority trial. Lancet. 2022;400(10349):380-390. doi:10.1016/S0140-6736(22)00916-3
Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet. 2011;377(9784):2181-2192. doi:10.1016/S0140-6736(11)60739-3
Ouchi Y, Sasaki J, Arai H, et al. Ezetimibe lipid-lowering trial on prevention of atherosclerotic cardiovascular disease in 75 or older (EWTOPIA 75): a randomized, controlled trial. Circulation. 2019;140:992-1003. doi:10.1161/CIRCULATIONAHA.118.039415
Baruch L, Gupta B, Lieberman-Blum SS, Agarwal S, Eng C. Ezetimibe 5 and 10 mg for lowering LDL-C: potential billion-dollar savings with improved tolerability. Am J Manag Care. 2008;14(10):637-641. https://www.ajmc.com/view/oct08-3644p637-641
Stroes ES, Thompson PD, Corsini A, et al. Statin-associated muscle symptoms: impact on statin therapy-European Atherosclerosis Society Consensus Panel Statement on Assessment, Aetiology and Management. Eur Heart J. 2015;36(17):1012-1022. doi:10.1093/eurheartj/ehv043
Stroes E, Colquhoun D, Sullivan D, et al. Anti-PCSK9 antibody effectively lowers cholesterol in patients with statin intolerance: the GAUSS-2 randomized, placebo-controlled phase 3 clinical trial of evolocumab. J Am Coll Cardiol. 2014;63(23):2541-2548. doi:10.1016/j.jacc.2014.03.019
Nissen SE, Stroes E, Dent-Acosta RE, et al. Efficacy and tolerability of evolocumab vs ezetimibe in patients with muscle-related statin intolerance: the GAUSS-3 randomized clinical trial. JAMA. 2016;315(15):1580-1590. doi:10.1001/jama.2016.3608
Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2019;73(24) e285-e350. doi:10.1016/j.jacc.2018.11.003
Writing Committee, Lloyd-Jones DM, Morris PB, et al. 2022 ACC expert consensus decision pathway on the role of nonstatin therapies for LDL-cholesterol lowering in the management of atherosclerotic cardiovascular disease risk: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2022;80(14):1366-1418. doi:10.1016/j.jacc.2022.07.006
US Food and Drug Administration. Ezetimibe. 2007. Accessed April 1, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/021445s019lbl.pdf
Singh A, Cho LS. Nonstatin therapy to reduce low-density lipoprotein cholesterol and improve cardiovascular outcomes. Cleve Clin J Med. 2024;91(1):53-63. doi:10.3949/ccjm.91a.23058
Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372(25):2387-2397. doi:10.1056/NEJMoa1410489
Kim B, Hong S, Lee Y, et al. Long-term efficacy and safety of moderate-intensity statin with ezetimibe combination therapy versus high-intensity statin monotherapy in patients with atherosclerotic cardiovascular disease (RACING): a randomised, open-label, non-inferiority trial. Lancet. 2022;400(10349):380-390. doi:10.1016/S0140-6736(22)00916-3
Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet. 2011;377(9784):2181-2192. doi:10.1016/S0140-6736(11)60739-3
Ouchi Y, Sasaki J, Arai H, et al. Ezetimibe lipid-lowering trial on prevention of atherosclerotic cardiovascular disease in 75 or older (EWTOPIA 75): a randomized, controlled trial. Circulation. 2019;140:992-1003. doi:10.1161/CIRCULATIONAHA.118.039415
Baruch L, Gupta B, Lieberman-Blum SS, Agarwal S, Eng C. Ezetimibe 5 and 10 mg for lowering LDL-C: potential billion-dollar savings with improved tolerability. Am J Manag Care. 2008;14(10):637-641. https://www.ajmc.com/view/oct08-3644p637-641
Stroes ES, Thompson PD, Corsini A, et al. Statin-associated muscle symptoms: impact on statin therapy-European Atherosclerosis Society Consensus Panel Statement on Assessment, Aetiology and Management. Eur Heart J. 2015;36(17):1012-1022. doi:10.1093/eurheartj/ehv043
Stroes E, Colquhoun D, Sullivan D, et al. Anti-PCSK9 antibody effectively lowers cholesterol in patients with statin intolerance: the GAUSS-2 randomized, placebo-controlled phase 3 clinical trial of evolocumab. J Am Coll Cardiol. 2014;63(23):2541-2548. doi:10.1016/j.jacc.2014.03.019
Nissen SE, Stroes E, Dent-Acosta RE, et al. Efficacy and tolerability of evolocumab vs ezetimibe in patients with muscle-related statin intolerance: the GAUSS-3 randomized clinical trial. JAMA. 2016;315(15):1580-1590. doi:10.1001/jama.2016.3608
Efficacy of Anti-Obesity Medications in Adult and Older Adult Veteran Populations
Efficacy of Anti-Obesity Medications in Adult and Older Adult Veteran Populations
The impact of obesity in the United States is significant. Between August 2021 and August 2023, the prevalence of obesity (body mass index ≥ 30) in US adults was 40.3%.1 The prevalence of obesity in adults aged 40 to 59 years was 46.4%, higher than the prevalence in adults aged 20 to 39 years (35.5%) and those aged ≥ 60 years (38.9%).1 The excess annual medical costs associated with obesity in the US are estimated at nearly $173 billion.2
The first-line treatment for obesity is lifestyle modifications, including a healthy diet and exercise. When lifestyle modifications are not enough to achieve weight-loss goals, bariatric surgery and anti-obesity medications (AOMs) are often considered. Five medications were approved for the long-term tretament of obesity by the US Food and Drug Administration (FDA) between 2021 and 2023, when this study was conducted: semaglutide (Wegovy), liraglutide (Saxenda), phentermine and topiramate, naltrexone and bupropion, and orlistat. The clinically meaningful (and commonly accepted) weight-loss target for these medications is ≥ 5% from baseline by week 12 of the maximally tolerated dose of therapy. A 5% weight loss has been shown to be clinically significant in improving cardiometabolic risk factors.3,4 These medications are intended to be used as an adjunct to healthy diet and exercise. Of note, semaglutide and liraglutide carry brand names, which are associated with different dosing for the treatment of type 2 diabetes mellitus (T2DM).
All 5 FDA-approved AOMs were available at the Veterans Affairs Sioux Falls Health Care System (VASFHCS) for the treatment of obesity at the time of the study. To qualify for an AOM, a veteran at VASFHCS must first work with a dietitian or be enrolled in the MOVE! clinic to participate in the weight management program, which focuses on dietary, exercise, and behavioral changes. At VASFHCS, AOMs are prescribed by primary care practitioners, clinical pharmacy providers, and advanced practitioners within the MOVE! program.
Ample data exist for the efficacy of AOMs. However, no published research has reported on AOM efficacy by age group (Appendix).5-11 While most of the AOM clinical trials included older adults, the average age of participants was typically between 40 and 50 years. It is well-known that pharmacokinetic and pharmacodynamic changes occur as age increases. Renal and hepatic clearance is reduced while the volume distribution and sensitivities to some medications may increase. 12 Although this study did not focus on specific pharmacokinetic and pharmacodynamic changes with respect to AOM, it is important to recognize that this may play a role in the efficacy and safety of AOMs in older adults.

Methods
This retrospective single-center chart review was performed using the VASFHCS Computerized Patient Record System to compare the efficacy of AOMs in older adults (aged ≥ 65 years) vs adults (aged < 65 years). The primary endpoint was the percent change in body weight from baseline to 6 and 12 months after initiation of AOM therapy in the older adult vs adult population. Secondary endpoints included changes in low-density lipoprotein (LDL), hemoglobin A1c (HbA1c), and blood pressure (BP) from baseline compared to 12 months on AOM therapy. HbA1c was assessed in patients with T2DM or prediabetes at the time of AOM initiation. Two safety endpoints were also explored to determine the incidence of medication adverse events (AEs) and subsequent discontinuation of AOM. A subset analysis was performed to determine whether there was a difference in percent change in body weight between patients in 3 age groups: 18 to 40 years, 41 to 64 years, and ≥ 65 years.
The study population included patients who were prescribed an AOM between January 1, 2021, and June 30, 2023. Patients were excluded if they did not continue AOM therapy for ≥ 6 months after initiation or if they underwent gastric bypass surgery while undergoing AOM therapy. Patients taking semaglutide (Ozempic) or liraglutide (Victoza) for both T2DM and weight loss who were eventually switched to the weight loss formulations (Wegovy or Saxenda) were included. Patients who switched between semaglutide and liraglutide for weight loss were also included. Those taking semaglutide or liraglutide solely for T2DM treatment were excluded because they are dosed differently.
Collected data included age, gender, race, weight (baseline, 6 and 12 months after initiation of AOM), metabolic laboratory values/vital signs (HbA1c, LDL, and BP at baseline and 12 months after initiation of AOM), diagnosis of T2DM or prediabetes, reported AEs associated with AOM therapy, and date of AOM initiation and discontinuation (if applicable). Baseline values were defined at the time of medication initiation or values documented within 6 months prior to medication initiation if true baseline data were not reported. If values were not recorded at months 6 and 12 after AOM initiation, values documented closest to those targets were used. Weights were used for baseline, 6-, and 12-month data unless they were unavailable due to use of virtual care modalities. In these cases, patient-reported weights were used. Patients were included in the 6-month data, but not the 12-month data, if they were taking AOMs for > 6 months but not for 12 months. If patients had been on multiple AOMs, baseline data were recorded at the start of the first medication that was used for 6 months or longer. Twelve-month data were recorded after subsequent medication change. Twelve-month metabolic laboratory values/vital signs were recorded for patients included in the study even if they did not complete ≥ 12 months of AOM therapy.
Statistical Analysis
Data from patients who were prescribed an AOM from January 2021 to June 2023 and who remained on the medication for ≥ 6 months were analyzed. Baseline characteristics were analyzed using descriptive statistics. The primary and secondary endpoints were evaluated using the t test. The safety endpoints were analyzed using descriptive statistics. An analysis of variance test was used for the subset analysis. Results with P < .05 were statistically significant.
Results
A total of 144 participants were included in this study, 116 in the adult group (aged < 65 years) and 28 in the older adult group (aged ≥ 65 years). Sixty-seven patients were excluded due to prespecified inclusion and exclusion criteria.
Other than the predetermined mean age differences (48 years vs 71 years), there were multiple differences in patient baseline characteristics. When comparing older adults and adults, average weight (283 lb vs 269 lb) and White race (89% vs 87%) were slightly higher in the older adult group. Also, a higher prevalence of T2DM (54% and 18%) and a lower prevalence of prediabetes (21% and 33%) was noted in the older adult group. HbA1c and BP were similar between both groups at baseline, while LDL was slightly lower in the older adult group (Table 1).

Patients in the adult group lost a mean 7.0% and 8.7% of body weight at 6 and 12 months, respectively, while the older adult group lost 5.0% and 6.6% body weight at 6 and 12 months, respectively. The difference in percent change in body weight was not statistically different at 6 (P = .08) or 12 (P = .26) months between patients in the adult group vs the older adult group or in the specific age groups (18-40 years, 41-64 years, ≥ 65 years) at 6 months (P = .24) or 12 months (P = .53) (Figure).

At 12 months, the difference between the adult group vs the older adult group was not statistically significant for HbA1c in patients with T2DM or prediabetes (P = .73), LDL (P = .95), systolic BP (P = .58), or diastolic BP (P = .51) (Table 2).

For the safety endpoint, the incidence of AEs was found to be different between groups. There were more reported AEs (61.2% vs 39.3%) and a greater increase in therapy discontinuation due to AEs (6.0% vs 0%) in the adult group compared to the older adult group (Table 3).

Discussion
Patients taking AOMs revealed no statistically significant difference in percent change in body weight at 6 or 12 months between adults aged < 65 years and older adults aged ≥ 65 years. The subset analysis also showed no statistically significant difference in change in percent body weight between more narrowly defined age groups of 18 to 40 years, 41 to 64 years, and ≥ 65 years. This suggests that AOM may have similar efficacy for weight loss in all ages of adults.
Secondary endpoint findings showed no statistically significant difference in HbA1c (in patients with T2DM or prediabetes), LDL, or BP at 12 months between the 2 groups. Although this study did not differentiate secondary outcomes based on the individual AOM, the change in HbA1c in both groups was expected, given that 70% of the patients included in this study were taking a glucagon-like peptide-1 agonist (liraglutide and semaglutide) at some point during the study. It’s also worth noting that secondary endpoints were collected for patients who discontinued the AOM between 6 and 12 months. Therefore, the patients’ HbA1c, LDL, and BP may not have accurately reflected the change that could have been expected if they had continued AOM therapy beyond the 12-month period.
Due to the different mechanisms and range in efficacy that AOMs have in regard to weight loss, changes in all outcomes, including weight, HbA1c, LDL, and BP were expected to vary as patients were included even after switching AOM (collection of data started after ≥ 6 months on a single AOM). Switching of AOM after the first 6 months of therapy was recorded in 25% of the patients in the ≥ 65 years group and 330% of the patients in the < 65 years group.
The incidence of AEs and subsequent discontinuation of AOMs in this study was higher in the adult group. This study excluded patients who did not continue taking an AOM for at least 6 months. As a result, the incidence of AEs between the 2 groups within the first 6 months of AOM therapy remains unknown. It is possible that during the first 6 months of therapy, patients aged < 65 years were more willing to tolerate or had fewer severe AEs compared with the older adult group. It’s also possible that the smaller number of patients in the older adult group was due to increased AEs that led them to discontinue early (before completion of 6 months of therapy) and/or prescriber discomfort in using AOMs in the older adult population. In addition, because the specific medication(s) taken by patients in each group were not detailed, it is unknown whether the adult group was taking AOMs associated with a greater number of AEs.
Limitations
This was a retrospective study with a relatively small sample size. A larger sample size may have shown more precise differences between age groups and may be more representative of the general population. Additionally, data were reliant on appropriate documentation, and adherence to AOM therapy was not assessed due to the retrospective nature of this study. At times, the study relied on patient reported data points, such as weight, if a clinic weight was not available. Also, this study did not account for many potential confounding factors such as other medications taken by the patient, which can affect outcomes including weight, HbA1c, LDL, blood pressure, and AEs.
Conclusions
This retrospective study of patients taking AOMs showed no statistically significant difference in weight loss at 6 or 12 months between adults aged < 65 years and older adults aged ≥ 65 years. A subset analysis found no statistically significant difference in change in body weight between specific age groups (18-40 years, 41-64 years, and ≥ 65 years). There was also no statistically significant difference in secondary outcomes, including change in HbA1c (in patients with T2DM or prediabetes), LDL or BP between age groups. The safety endpoints showed a higher incidence of medication AEs in the adult group, with more of these adults discontinuing therapy due to AEs. This study indicates that AOM may have similar outcomes for weight loss and metabolic laboratory values/vital sign changes between adults and older adults. Also, our findings suggest that patients aged < 65 years may experience more AEs than patients aged ≥ 65 years after ≥ 6 months of AOM therapy. Larger studies are needed to further evaluate these age-specific findings.
- Emmerich SD, Fryar CD, Stierman B, Ogden CL. Obesity and severe obesity prevalence in adults: United States, August 2021-August 2023. NCHS Data Brief No. 508. National Center for Health Statistics; 2024. Accessed December 11, 2024. https://www.cdc.gov/nchs/products/databriefs/db508.htm
- Ward ZJ, Bleich SN, Long MW, Gortmaker SL. Association of body mass index with health care expenditures in the United States by age and sex. PLoS One. 2021;16(3):e0247307. doi:10.1371/journal.pone.0247307
- Horn DB, Almandoz JP, Look M. What is clinically relevant weight loss for your patients and how can it be achieved? A narrative review. Postgrad Med. 2022;134(4):359-375. doi:10.1080/00325481.2022.2051366
- American Diabetes Association (ADA). Standards of care in diabetes–2023. Diabetes Care. 2023;46(suppl 1):S128- S2139. doi:10.2337/dc23-S008
- Wilding JPH, Batterham RL, Calanna S, et al. Onceweekly semaglutide in adults with overweight or obesity. N Engl J Med. 2021;384(11):989-1002. doi:10.1056/NEJMoa2032183
- Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med. 2015;373(1):11-22. doi:10.1056/NEJMoa1411892
- Allison DB, Gadde KM, Garvey WT, et al. Controlled-release phentermine/topiramate in severely obese adults: a randomized controlled trial (EQUIP). Obesity (Silver Spring). 2012;20(2):330-342. doi:10.1038/oby.2011.330
- Gadde KM, Allison DB, Ryan DH, et al. Effects of low-dose, controlled-release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377(9774):1341-1352. doi:10.1016/S0140-6736(11)60205-5
- Garvey WT, Ryan DH, Look M, et al. Two-year sustained weight loss and metabolic benefits with controlled-release phentermine/topiramate in obese and overweight adults (SEQUEL): a randomized, placebo-controlled, phase 3 extension study. Am J Clin Nutr. 2012;95(2):297-308. doi:10.3945/ajcn.111.024927
- Greenway FL, Fujioka K, Plodkowski RA, et al. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2010;376(9741):595-605. doi:10.1016/S0140-6736(10)60888-4
- Sjöström L, Rissanen A, Andersen T, et al. Randomised placebo-controlled trial of orlistat for weight loss and prevention of weight regain in obese patients. European Multicentre Orlistat Study Group. Lancet. 1998;352(9123):167-172. doi:10.1016s0140-6736(97)11509-4
- Mangoni AA, Jackson SHD. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6-14. doi:10.1046/j.1365-2125.2003.02007.x
The impact of obesity in the United States is significant. Between August 2021 and August 2023, the prevalence of obesity (body mass index ≥ 30) in US adults was 40.3%.1 The prevalence of obesity in adults aged 40 to 59 years was 46.4%, higher than the prevalence in adults aged 20 to 39 years (35.5%) and those aged ≥ 60 years (38.9%).1 The excess annual medical costs associated with obesity in the US are estimated at nearly $173 billion.2
The first-line treatment for obesity is lifestyle modifications, including a healthy diet and exercise. When lifestyle modifications are not enough to achieve weight-loss goals, bariatric surgery and anti-obesity medications (AOMs) are often considered. Five medications were approved for the long-term tretament of obesity by the US Food and Drug Administration (FDA) between 2021 and 2023, when this study was conducted: semaglutide (Wegovy), liraglutide (Saxenda), phentermine and topiramate, naltrexone and bupropion, and orlistat. The clinically meaningful (and commonly accepted) weight-loss target for these medications is ≥ 5% from baseline by week 12 of the maximally tolerated dose of therapy. A 5% weight loss has been shown to be clinically significant in improving cardiometabolic risk factors.3,4 These medications are intended to be used as an adjunct to healthy diet and exercise. Of note, semaglutide and liraglutide carry brand names, which are associated with different dosing for the treatment of type 2 diabetes mellitus (T2DM).
All 5 FDA-approved AOMs were available at the Veterans Affairs Sioux Falls Health Care System (VASFHCS) for the treatment of obesity at the time of the study. To qualify for an AOM, a veteran at VASFHCS must first work with a dietitian or be enrolled in the MOVE! clinic to participate in the weight management program, which focuses on dietary, exercise, and behavioral changes. At VASFHCS, AOMs are prescribed by primary care practitioners, clinical pharmacy providers, and advanced practitioners within the MOVE! program.
Ample data exist for the efficacy of AOMs. However, no published research has reported on AOM efficacy by age group (Appendix).5-11 While most of the AOM clinical trials included older adults, the average age of participants was typically between 40 and 50 years. It is well-known that pharmacokinetic and pharmacodynamic changes occur as age increases. Renal and hepatic clearance is reduced while the volume distribution and sensitivities to some medications may increase. 12 Although this study did not focus on specific pharmacokinetic and pharmacodynamic changes with respect to AOM, it is important to recognize that this may play a role in the efficacy and safety of AOMs in older adults.

Methods
This retrospective single-center chart review was performed using the VASFHCS Computerized Patient Record System to compare the efficacy of AOMs in older adults (aged ≥ 65 years) vs adults (aged < 65 years). The primary endpoint was the percent change in body weight from baseline to 6 and 12 months after initiation of AOM therapy in the older adult vs adult population. Secondary endpoints included changes in low-density lipoprotein (LDL), hemoglobin A1c (HbA1c), and blood pressure (BP) from baseline compared to 12 months on AOM therapy. HbA1c was assessed in patients with T2DM or prediabetes at the time of AOM initiation. Two safety endpoints were also explored to determine the incidence of medication adverse events (AEs) and subsequent discontinuation of AOM. A subset analysis was performed to determine whether there was a difference in percent change in body weight between patients in 3 age groups: 18 to 40 years, 41 to 64 years, and ≥ 65 years.
The study population included patients who were prescribed an AOM between January 1, 2021, and June 30, 2023. Patients were excluded if they did not continue AOM therapy for ≥ 6 months after initiation or if they underwent gastric bypass surgery while undergoing AOM therapy. Patients taking semaglutide (Ozempic) or liraglutide (Victoza) for both T2DM and weight loss who were eventually switched to the weight loss formulations (Wegovy or Saxenda) were included. Patients who switched between semaglutide and liraglutide for weight loss were also included. Those taking semaglutide or liraglutide solely for T2DM treatment were excluded because they are dosed differently.
Collected data included age, gender, race, weight (baseline, 6 and 12 months after initiation of AOM), metabolic laboratory values/vital signs (HbA1c, LDL, and BP at baseline and 12 months after initiation of AOM), diagnosis of T2DM or prediabetes, reported AEs associated with AOM therapy, and date of AOM initiation and discontinuation (if applicable). Baseline values were defined at the time of medication initiation or values documented within 6 months prior to medication initiation if true baseline data were not reported. If values were not recorded at months 6 and 12 after AOM initiation, values documented closest to those targets were used. Weights were used for baseline, 6-, and 12-month data unless they were unavailable due to use of virtual care modalities. In these cases, patient-reported weights were used. Patients were included in the 6-month data, but not the 12-month data, if they were taking AOMs for > 6 months but not for 12 months. If patients had been on multiple AOMs, baseline data were recorded at the start of the first medication that was used for 6 months or longer. Twelve-month data were recorded after subsequent medication change. Twelve-month metabolic laboratory values/vital signs were recorded for patients included in the study even if they did not complete ≥ 12 months of AOM therapy.
Statistical Analysis
Data from patients who were prescribed an AOM from January 2021 to June 2023 and who remained on the medication for ≥ 6 months were analyzed. Baseline characteristics were analyzed using descriptive statistics. The primary and secondary endpoints were evaluated using the t test. The safety endpoints were analyzed using descriptive statistics. An analysis of variance test was used for the subset analysis. Results with P < .05 were statistically significant.
Results
A total of 144 participants were included in this study, 116 in the adult group (aged < 65 years) and 28 in the older adult group (aged ≥ 65 years). Sixty-seven patients were excluded due to prespecified inclusion and exclusion criteria.
Other than the predetermined mean age differences (48 years vs 71 years), there were multiple differences in patient baseline characteristics. When comparing older adults and adults, average weight (283 lb vs 269 lb) and White race (89% vs 87%) were slightly higher in the older adult group. Also, a higher prevalence of T2DM (54% and 18%) and a lower prevalence of prediabetes (21% and 33%) was noted in the older adult group. HbA1c and BP were similar between both groups at baseline, while LDL was slightly lower in the older adult group (Table 1).

Patients in the adult group lost a mean 7.0% and 8.7% of body weight at 6 and 12 months, respectively, while the older adult group lost 5.0% and 6.6% body weight at 6 and 12 months, respectively. The difference in percent change in body weight was not statistically different at 6 (P = .08) or 12 (P = .26) months between patients in the adult group vs the older adult group or in the specific age groups (18-40 years, 41-64 years, ≥ 65 years) at 6 months (P = .24) or 12 months (P = .53) (Figure).

At 12 months, the difference between the adult group vs the older adult group was not statistically significant for HbA1c in patients with T2DM or prediabetes (P = .73), LDL (P = .95), systolic BP (P = .58), or diastolic BP (P = .51) (Table 2).

For the safety endpoint, the incidence of AEs was found to be different between groups. There were more reported AEs (61.2% vs 39.3%) and a greater increase in therapy discontinuation due to AEs (6.0% vs 0%) in the adult group compared to the older adult group (Table 3).

Discussion
Patients taking AOMs revealed no statistically significant difference in percent change in body weight at 6 or 12 months between adults aged < 65 years and older adults aged ≥ 65 years. The subset analysis also showed no statistically significant difference in change in percent body weight between more narrowly defined age groups of 18 to 40 years, 41 to 64 years, and ≥ 65 years. This suggests that AOM may have similar efficacy for weight loss in all ages of adults.
Secondary endpoint findings showed no statistically significant difference in HbA1c (in patients with T2DM or prediabetes), LDL, or BP at 12 months between the 2 groups. Although this study did not differentiate secondary outcomes based on the individual AOM, the change in HbA1c in both groups was expected, given that 70% of the patients included in this study were taking a glucagon-like peptide-1 agonist (liraglutide and semaglutide) at some point during the study. It’s also worth noting that secondary endpoints were collected for patients who discontinued the AOM between 6 and 12 months. Therefore, the patients’ HbA1c, LDL, and BP may not have accurately reflected the change that could have been expected if they had continued AOM therapy beyond the 12-month period.
Due to the different mechanisms and range in efficacy that AOMs have in regard to weight loss, changes in all outcomes, including weight, HbA1c, LDL, and BP were expected to vary as patients were included even after switching AOM (collection of data started after ≥ 6 months on a single AOM). Switching of AOM after the first 6 months of therapy was recorded in 25% of the patients in the ≥ 65 years group and 330% of the patients in the < 65 years group.
The incidence of AEs and subsequent discontinuation of AOMs in this study was higher in the adult group. This study excluded patients who did not continue taking an AOM for at least 6 months. As a result, the incidence of AEs between the 2 groups within the first 6 months of AOM therapy remains unknown. It is possible that during the first 6 months of therapy, patients aged < 65 years were more willing to tolerate or had fewer severe AEs compared with the older adult group. It’s also possible that the smaller number of patients in the older adult group was due to increased AEs that led them to discontinue early (before completion of 6 months of therapy) and/or prescriber discomfort in using AOMs in the older adult population. In addition, because the specific medication(s) taken by patients in each group were not detailed, it is unknown whether the adult group was taking AOMs associated with a greater number of AEs.
Limitations
This was a retrospective study with a relatively small sample size. A larger sample size may have shown more precise differences between age groups and may be more representative of the general population. Additionally, data were reliant on appropriate documentation, and adherence to AOM therapy was not assessed due to the retrospective nature of this study. At times, the study relied on patient reported data points, such as weight, if a clinic weight was not available. Also, this study did not account for many potential confounding factors such as other medications taken by the patient, which can affect outcomes including weight, HbA1c, LDL, blood pressure, and AEs.
Conclusions
This retrospective study of patients taking AOMs showed no statistically significant difference in weight loss at 6 or 12 months between adults aged < 65 years and older adults aged ≥ 65 years. A subset analysis found no statistically significant difference in change in body weight between specific age groups (18-40 years, 41-64 years, and ≥ 65 years). There was also no statistically significant difference in secondary outcomes, including change in HbA1c (in patients with T2DM or prediabetes), LDL or BP between age groups. The safety endpoints showed a higher incidence of medication AEs in the adult group, with more of these adults discontinuing therapy due to AEs. This study indicates that AOM may have similar outcomes for weight loss and metabolic laboratory values/vital sign changes between adults and older adults. Also, our findings suggest that patients aged < 65 years may experience more AEs than patients aged ≥ 65 years after ≥ 6 months of AOM therapy. Larger studies are needed to further evaluate these age-specific findings.
The impact of obesity in the United States is significant. Between August 2021 and August 2023, the prevalence of obesity (body mass index ≥ 30) in US adults was 40.3%.1 The prevalence of obesity in adults aged 40 to 59 years was 46.4%, higher than the prevalence in adults aged 20 to 39 years (35.5%) and those aged ≥ 60 years (38.9%).1 The excess annual medical costs associated with obesity in the US are estimated at nearly $173 billion.2
The first-line treatment for obesity is lifestyle modifications, including a healthy diet and exercise. When lifestyle modifications are not enough to achieve weight-loss goals, bariatric surgery and anti-obesity medications (AOMs) are often considered. Five medications were approved for the long-term tretament of obesity by the US Food and Drug Administration (FDA) between 2021 and 2023, when this study was conducted: semaglutide (Wegovy), liraglutide (Saxenda), phentermine and topiramate, naltrexone and bupropion, and orlistat. The clinically meaningful (and commonly accepted) weight-loss target for these medications is ≥ 5% from baseline by week 12 of the maximally tolerated dose of therapy. A 5% weight loss has been shown to be clinically significant in improving cardiometabolic risk factors.3,4 These medications are intended to be used as an adjunct to healthy diet and exercise. Of note, semaglutide and liraglutide carry brand names, which are associated with different dosing for the treatment of type 2 diabetes mellitus (T2DM).
All 5 FDA-approved AOMs were available at the Veterans Affairs Sioux Falls Health Care System (VASFHCS) for the treatment of obesity at the time of the study. To qualify for an AOM, a veteran at VASFHCS must first work with a dietitian or be enrolled in the MOVE! clinic to participate in the weight management program, which focuses on dietary, exercise, and behavioral changes. At VASFHCS, AOMs are prescribed by primary care practitioners, clinical pharmacy providers, and advanced practitioners within the MOVE! program.
Ample data exist for the efficacy of AOMs. However, no published research has reported on AOM efficacy by age group (Appendix).5-11 While most of the AOM clinical trials included older adults, the average age of participants was typically between 40 and 50 years. It is well-known that pharmacokinetic and pharmacodynamic changes occur as age increases. Renal and hepatic clearance is reduced while the volume distribution and sensitivities to some medications may increase. 12 Although this study did not focus on specific pharmacokinetic and pharmacodynamic changes with respect to AOM, it is important to recognize that this may play a role in the efficacy and safety of AOMs in older adults.

Methods
This retrospective single-center chart review was performed using the VASFHCS Computerized Patient Record System to compare the efficacy of AOMs in older adults (aged ≥ 65 years) vs adults (aged < 65 years). The primary endpoint was the percent change in body weight from baseline to 6 and 12 months after initiation of AOM therapy in the older adult vs adult population. Secondary endpoints included changes in low-density lipoprotein (LDL), hemoglobin A1c (HbA1c), and blood pressure (BP) from baseline compared to 12 months on AOM therapy. HbA1c was assessed in patients with T2DM or prediabetes at the time of AOM initiation. Two safety endpoints were also explored to determine the incidence of medication adverse events (AEs) and subsequent discontinuation of AOM. A subset analysis was performed to determine whether there was a difference in percent change in body weight between patients in 3 age groups: 18 to 40 years, 41 to 64 years, and ≥ 65 years.
The study population included patients who were prescribed an AOM between January 1, 2021, and June 30, 2023. Patients were excluded if they did not continue AOM therapy for ≥ 6 months after initiation or if they underwent gastric bypass surgery while undergoing AOM therapy. Patients taking semaglutide (Ozempic) or liraglutide (Victoza) for both T2DM and weight loss who were eventually switched to the weight loss formulations (Wegovy or Saxenda) were included. Patients who switched between semaglutide and liraglutide for weight loss were also included. Those taking semaglutide or liraglutide solely for T2DM treatment were excluded because they are dosed differently.
Collected data included age, gender, race, weight (baseline, 6 and 12 months after initiation of AOM), metabolic laboratory values/vital signs (HbA1c, LDL, and BP at baseline and 12 months after initiation of AOM), diagnosis of T2DM or prediabetes, reported AEs associated with AOM therapy, and date of AOM initiation and discontinuation (if applicable). Baseline values were defined at the time of medication initiation or values documented within 6 months prior to medication initiation if true baseline data were not reported. If values were not recorded at months 6 and 12 after AOM initiation, values documented closest to those targets were used. Weights were used for baseline, 6-, and 12-month data unless they were unavailable due to use of virtual care modalities. In these cases, patient-reported weights were used. Patients were included in the 6-month data, but not the 12-month data, if they were taking AOMs for > 6 months but not for 12 months. If patients had been on multiple AOMs, baseline data were recorded at the start of the first medication that was used for 6 months or longer. Twelve-month data were recorded after subsequent medication change. Twelve-month metabolic laboratory values/vital signs were recorded for patients included in the study even if they did not complete ≥ 12 months of AOM therapy.
Statistical Analysis
Data from patients who were prescribed an AOM from January 2021 to June 2023 and who remained on the medication for ≥ 6 months were analyzed. Baseline characteristics were analyzed using descriptive statistics. The primary and secondary endpoints were evaluated using the t test. The safety endpoints were analyzed using descriptive statistics. An analysis of variance test was used for the subset analysis. Results with P < .05 were statistically significant.
Results
A total of 144 participants were included in this study, 116 in the adult group (aged < 65 years) and 28 in the older adult group (aged ≥ 65 years). Sixty-seven patients were excluded due to prespecified inclusion and exclusion criteria.
Other than the predetermined mean age differences (48 years vs 71 years), there were multiple differences in patient baseline characteristics. When comparing older adults and adults, average weight (283 lb vs 269 lb) and White race (89% vs 87%) were slightly higher in the older adult group. Also, a higher prevalence of T2DM (54% and 18%) and a lower prevalence of prediabetes (21% and 33%) was noted in the older adult group. HbA1c and BP were similar between both groups at baseline, while LDL was slightly lower in the older adult group (Table 1).

Patients in the adult group lost a mean 7.0% and 8.7% of body weight at 6 and 12 months, respectively, while the older adult group lost 5.0% and 6.6% body weight at 6 and 12 months, respectively. The difference in percent change in body weight was not statistically different at 6 (P = .08) or 12 (P = .26) months between patients in the adult group vs the older adult group or in the specific age groups (18-40 years, 41-64 years, ≥ 65 years) at 6 months (P = .24) or 12 months (P = .53) (Figure).

At 12 months, the difference between the adult group vs the older adult group was not statistically significant for HbA1c in patients with T2DM or prediabetes (P = .73), LDL (P = .95), systolic BP (P = .58), or diastolic BP (P = .51) (Table 2).

For the safety endpoint, the incidence of AEs was found to be different between groups. There were more reported AEs (61.2% vs 39.3%) and a greater increase in therapy discontinuation due to AEs (6.0% vs 0%) in the adult group compared to the older adult group (Table 3).

Discussion
Patients taking AOMs revealed no statistically significant difference in percent change in body weight at 6 or 12 months between adults aged < 65 years and older adults aged ≥ 65 years. The subset analysis also showed no statistically significant difference in change in percent body weight between more narrowly defined age groups of 18 to 40 years, 41 to 64 years, and ≥ 65 years. This suggests that AOM may have similar efficacy for weight loss in all ages of adults.
Secondary endpoint findings showed no statistically significant difference in HbA1c (in patients with T2DM or prediabetes), LDL, or BP at 12 months between the 2 groups. Although this study did not differentiate secondary outcomes based on the individual AOM, the change in HbA1c in both groups was expected, given that 70% of the patients included in this study were taking a glucagon-like peptide-1 agonist (liraglutide and semaglutide) at some point during the study. It’s also worth noting that secondary endpoints were collected for patients who discontinued the AOM between 6 and 12 months. Therefore, the patients’ HbA1c, LDL, and BP may not have accurately reflected the change that could have been expected if they had continued AOM therapy beyond the 12-month period.
Due to the different mechanisms and range in efficacy that AOMs have in regard to weight loss, changes in all outcomes, including weight, HbA1c, LDL, and BP were expected to vary as patients were included even after switching AOM (collection of data started after ≥ 6 months on a single AOM). Switching of AOM after the first 6 months of therapy was recorded in 25% of the patients in the ≥ 65 years group and 330% of the patients in the < 65 years group.
The incidence of AEs and subsequent discontinuation of AOMs in this study was higher in the adult group. This study excluded patients who did not continue taking an AOM for at least 6 months. As a result, the incidence of AEs between the 2 groups within the first 6 months of AOM therapy remains unknown. It is possible that during the first 6 months of therapy, patients aged < 65 years were more willing to tolerate or had fewer severe AEs compared with the older adult group. It’s also possible that the smaller number of patients in the older adult group was due to increased AEs that led them to discontinue early (before completion of 6 months of therapy) and/or prescriber discomfort in using AOMs in the older adult population. In addition, because the specific medication(s) taken by patients in each group were not detailed, it is unknown whether the adult group was taking AOMs associated with a greater number of AEs.
Limitations
This was a retrospective study with a relatively small sample size. A larger sample size may have shown more precise differences between age groups and may be more representative of the general population. Additionally, data were reliant on appropriate documentation, and adherence to AOM therapy was not assessed due to the retrospective nature of this study. At times, the study relied on patient reported data points, such as weight, if a clinic weight was not available. Also, this study did not account for many potential confounding factors such as other medications taken by the patient, which can affect outcomes including weight, HbA1c, LDL, blood pressure, and AEs.
Conclusions
This retrospective study of patients taking AOMs showed no statistically significant difference in weight loss at 6 or 12 months between adults aged < 65 years and older adults aged ≥ 65 years. A subset analysis found no statistically significant difference in change in body weight between specific age groups (18-40 years, 41-64 years, and ≥ 65 years). There was also no statistically significant difference in secondary outcomes, including change in HbA1c (in patients with T2DM or prediabetes), LDL or BP between age groups. The safety endpoints showed a higher incidence of medication AEs in the adult group, with more of these adults discontinuing therapy due to AEs. This study indicates that AOM may have similar outcomes for weight loss and metabolic laboratory values/vital sign changes between adults and older adults. Also, our findings suggest that patients aged < 65 years may experience more AEs than patients aged ≥ 65 years after ≥ 6 months of AOM therapy. Larger studies are needed to further evaluate these age-specific findings.
- Emmerich SD, Fryar CD, Stierman B, Ogden CL. Obesity and severe obesity prevalence in adults: United States, August 2021-August 2023. NCHS Data Brief No. 508. National Center for Health Statistics; 2024. Accessed December 11, 2024. https://www.cdc.gov/nchs/products/databriefs/db508.htm
- Ward ZJ, Bleich SN, Long MW, Gortmaker SL. Association of body mass index with health care expenditures in the United States by age and sex. PLoS One. 2021;16(3):e0247307. doi:10.1371/journal.pone.0247307
- Horn DB, Almandoz JP, Look M. What is clinically relevant weight loss for your patients and how can it be achieved? A narrative review. Postgrad Med. 2022;134(4):359-375. doi:10.1080/00325481.2022.2051366
- American Diabetes Association (ADA). Standards of care in diabetes–2023. Diabetes Care. 2023;46(suppl 1):S128- S2139. doi:10.2337/dc23-S008
- Wilding JPH, Batterham RL, Calanna S, et al. Onceweekly semaglutide in adults with overweight or obesity. N Engl J Med. 2021;384(11):989-1002. doi:10.1056/NEJMoa2032183
- Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med. 2015;373(1):11-22. doi:10.1056/NEJMoa1411892
- Allison DB, Gadde KM, Garvey WT, et al. Controlled-release phentermine/topiramate in severely obese adults: a randomized controlled trial (EQUIP). Obesity (Silver Spring). 2012;20(2):330-342. doi:10.1038/oby.2011.330
- Gadde KM, Allison DB, Ryan DH, et al. Effects of low-dose, controlled-release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377(9774):1341-1352. doi:10.1016/S0140-6736(11)60205-5
- Garvey WT, Ryan DH, Look M, et al. Two-year sustained weight loss and metabolic benefits with controlled-release phentermine/topiramate in obese and overweight adults (SEQUEL): a randomized, placebo-controlled, phase 3 extension study. Am J Clin Nutr. 2012;95(2):297-308. doi:10.3945/ajcn.111.024927
- Greenway FL, Fujioka K, Plodkowski RA, et al. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2010;376(9741):595-605. doi:10.1016/S0140-6736(10)60888-4
- Sjöström L, Rissanen A, Andersen T, et al. Randomised placebo-controlled trial of orlistat for weight loss and prevention of weight regain in obese patients. European Multicentre Orlistat Study Group. Lancet. 1998;352(9123):167-172. doi:10.1016s0140-6736(97)11509-4
- Mangoni AA, Jackson SHD. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6-14. doi:10.1046/j.1365-2125.2003.02007.x
- Emmerich SD, Fryar CD, Stierman B, Ogden CL. Obesity and severe obesity prevalence in adults: United States, August 2021-August 2023. NCHS Data Brief No. 508. National Center for Health Statistics; 2024. Accessed December 11, 2024. https://www.cdc.gov/nchs/products/databriefs/db508.htm
- Ward ZJ, Bleich SN, Long MW, Gortmaker SL. Association of body mass index with health care expenditures in the United States by age and sex. PLoS One. 2021;16(3):e0247307. doi:10.1371/journal.pone.0247307
- Horn DB, Almandoz JP, Look M. What is clinically relevant weight loss for your patients and how can it be achieved? A narrative review. Postgrad Med. 2022;134(4):359-375. doi:10.1080/00325481.2022.2051366
- American Diabetes Association (ADA). Standards of care in diabetes–2023. Diabetes Care. 2023;46(suppl 1):S128- S2139. doi:10.2337/dc23-S008
- Wilding JPH, Batterham RL, Calanna S, et al. Onceweekly semaglutide in adults with overweight or obesity. N Engl J Med. 2021;384(11):989-1002. doi:10.1056/NEJMoa2032183
- Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med. 2015;373(1):11-22. doi:10.1056/NEJMoa1411892
- Allison DB, Gadde KM, Garvey WT, et al. Controlled-release phentermine/topiramate in severely obese adults: a randomized controlled trial (EQUIP). Obesity (Silver Spring). 2012;20(2):330-342. doi:10.1038/oby.2011.330
- Gadde KM, Allison DB, Ryan DH, et al. Effects of low-dose, controlled-release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377(9774):1341-1352. doi:10.1016/S0140-6736(11)60205-5
- Garvey WT, Ryan DH, Look M, et al. Two-year sustained weight loss and metabolic benefits with controlled-release phentermine/topiramate in obese and overweight adults (SEQUEL): a randomized, placebo-controlled, phase 3 extension study. Am J Clin Nutr. 2012;95(2):297-308. doi:10.3945/ajcn.111.024927
- Greenway FL, Fujioka K, Plodkowski RA, et al. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2010;376(9741):595-605. doi:10.1016/S0140-6736(10)60888-4
- Sjöström L, Rissanen A, Andersen T, et al. Randomised placebo-controlled trial of orlistat for weight loss and prevention of weight regain in obese patients. European Multicentre Orlistat Study Group. Lancet. 1998;352(9123):167-172. doi:10.1016s0140-6736(97)11509-4
- Mangoni AA, Jackson SHD. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6-14. doi:10.1046/j.1365-2125.2003.02007.x
Efficacy of Anti-Obesity Medications in Adult and Older Adult Veteran Populations
Efficacy of Anti-Obesity Medications in Adult and Older Adult Veteran Populations
FDA Approves Sotorasib + Panitumumab for mCRC
The US Food and Drug Administration (FDA) has approved sotorasib (Lumakras, Amgen Inc.) with panitumumab (Vectibix, Amgen Inc.) for the treatment of certain adult patients with metastatic colorectal cancer (mCRC).
Specifically, the combination therapy is indicated for those with KRAS G12C-mutated mCRC, as determined using an FDA-approved test, who have received prior treatment with fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy, according to the FDA notice. The FDA also approved the therascreen KRAS RGQ PCR Kit (QIAGEN GmbH) as a companion diagnostic device for identifying eligible patients.
Approval of sotorasib with panitumumab was based on findings from the randomized, open-label, controlled CodeBreaK 300 trial showing improved overall response rates (ORR) and progression-free survival (PFS) with sotorasib and panitumumab vs investigator’s choice of trifluridine/tipiracil or regorafenib, which are current standard-of-care options.
Median PFS was 5.6 months in 53 patients randomized to receive 960 mg of oral sotorasib once daily plus 6 mg/kg of intravenous (IV) panitumumab every 2 weeks, and 2 months in 54 patients randomized to receive standard-of-care therapy (hazard ratio, 0.48). The ORR was 26% vs 0% in the arms, respectively, and the duration of response in the sotorasib/panitumumab arm was 4.4 months. No significant difference in PFS was observed between the standard-of-care arm and a third arm with 53 patients who received 240 mg of oral sotorasib daily plus 6 mg/kg of IV panitumumab every 2 weeks.
Overall survival (OS) did not differ significantly between the treatment arms in the final analysis, but the study was not statistically powered for OS.
Adverse reactions occurring in at least 20% of patients receiving sotorasib/panitumumab were rash, dry skin, diarrhea, stomatitis, fatigue, and musculoskeletal pain. Common grade 3-4 laboratory abnormalities, which occurred in two or more patients, included decreased magnesium, decreased potassium, decreased corrected calcium, and increased potassium.
The recommended dose of sotorasib is 960 mg given orally once daily and administered before the first panitumumab infusion. The recommended panitumumab dose is 6 mg/kg as an IV infusion every 14 days until disease progression, unacceptable toxicity, or until sotorasib is withheld or discontinued, according to the full prescribing information.
A version of this article first appeared on Medscape.com.
The US Food and Drug Administration (FDA) has approved sotorasib (Lumakras, Amgen Inc.) with panitumumab (Vectibix, Amgen Inc.) for the treatment of certain adult patients with metastatic colorectal cancer (mCRC).
Specifically, the combination therapy is indicated for those with KRAS G12C-mutated mCRC, as determined using an FDA-approved test, who have received prior treatment with fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy, according to the FDA notice. The FDA also approved the therascreen KRAS RGQ PCR Kit (QIAGEN GmbH) as a companion diagnostic device for identifying eligible patients.
Approval of sotorasib with panitumumab was based on findings from the randomized, open-label, controlled CodeBreaK 300 trial showing improved overall response rates (ORR) and progression-free survival (PFS) with sotorasib and panitumumab vs investigator’s choice of trifluridine/tipiracil or regorafenib, which are current standard-of-care options.
Median PFS was 5.6 months in 53 patients randomized to receive 960 mg of oral sotorasib once daily plus 6 mg/kg of intravenous (IV) panitumumab every 2 weeks, and 2 months in 54 patients randomized to receive standard-of-care therapy (hazard ratio, 0.48). The ORR was 26% vs 0% in the arms, respectively, and the duration of response in the sotorasib/panitumumab arm was 4.4 months. No significant difference in PFS was observed between the standard-of-care arm and a third arm with 53 patients who received 240 mg of oral sotorasib daily plus 6 mg/kg of IV panitumumab every 2 weeks.
Overall survival (OS) did not differ significantly between the treatment arms in the final analysis, but the study was not statistically powered for OS.
Adverse reactions occurring in at least 20% of patients receiving sotorasib/panitumumab were rash, dry skin, diarrhea, stomatitis, fatigue, and musculoskeletal pain. Common grade 3-4 laboratory abnormalities, which occurred in two or more patients, included decreased magnesium, decreased potassium, decreased corrected calcium, and increased potassium.
The recommended dose of sotorasib is 960 mg given orally once daily and administered before the first panitumumab infusion. The recommended panitumumab dose is 6 mg/kg as an IV infusion every 14 days until disease progression, unacceptable toxicity, or until sotorasib is withheld or discontinued, according to the full prescribing information.
A version of this article first appeared on Medscape.com.
The US Food and Drug Administration (FDA) has approved sotorasib (Lumakras, Amgen Inc.) with panitumumab (Vectibix, Amgen Inc.) for the treatment of certain adult patients with metastatic colorectal cancer (mCRC).
Specifically, the combination therapy is indicated for those with KRAS G12C-mutated mCRC, as determined using an FDA-approved test, who have received prior treatment with fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy, according to the FDA notice. The FDA also approved the therascreen KRAS RGQ PCR Kit (QIAGEN GmbH) as a companion diagnostic device for identifying eligible patients.
Approval of sotorasib with panitumumab was based on findings from the randomized, open-label, controlled CodeBreaK 300 trial showing improved overall response rates (ORR) and progression-free survival (PFS) with sotorasib and panitumumab vs investigator’s choice of trifluridine/tipiracil or regorafenib, which are current standard-of-care options.
Median PFS was 5.6 months in 53 patients randomized to receive 960 mg of oral sotorasib once daily plus 6 mg/kg of intravenous (IV) panitumumab every 2 weeks, and 2 months in 54 patients randomized to receive standard-of-care therapy (hazard ratio, 0.48). The ORR was 26% vs 0% in the arms, respectively, and the duration of response in the sotorasib/panitumumab arm was 4.4 months. No significant difference in PFS was observed between the standard-of-care arm and a third arm with 53 patients who received 240 mg of oral sotorasib daily plus 6 mg/kg of IV panitumumab every 2 weeks.
Overall survival (OS) did not differ significantly between the treatment arms in the final analysis, but the study was not statistically powered for OS.
Adverse reactions occurring in at least 20% of patients receiving sotorasib/panitumumab were rash, dry skin, diarrhea, stomatitis, fatigue, and musculoskeletal pain. Common grade 3-4 laboratory abnormalities, which occurred in two or more patients, included decreased magnesium, decreased potassium, decreased corrected calcium, and increased potassium.
The recommended dose of sotorasib is 960 mg given orally once daily and administered before the first panitumumab infusion. The recommended panitumumab dose is 6 mg/kg as an IV infusion every 14 days until disease progression, unacceptable toxicity, or until sotorasib is withheld or discontinued, according to the full prescribing information.
A version of this article first appeared on Medscape.com.
Pharmacist-Led Deprescribing of Aspirin for Primary Prevention of Cardiovascular Disease Among Geriatric Veterans
Pharmacist-Led Deprescribing of Aspirin for Primary Prevention of Cardiovascular Disease Among Geriatric Veterans
Low-dose aspirin commonly is used for the prevention of cardiovascular disease (CVD) but is associated with an increased risk of major bleeding.1 The use of aspirin for primary prevention is largely extrapolated from clinical trials showing benefit in the secondary prevention of myocardial infarction and ischemic stroke. However, results from the Aspirin in Reducing Events in the Elderly (ASPREE) trial challenged this practice.2 The ASPREE trial, conducted in the United States and Australia from 2010 to 2014, sought to determine whether daily 100 mg aspirin, was superior to placebo in promoting disability-free survival among older adults. Participants were aged ≥ 70 years (≥ 65 years for Hispanic and Black US participants), living in the community, and were free from preexisting CVD, cerebrovascular disease, or any chronic condition likely to limit survival to < 5 years. The study found no significant difference in the primary endpoints of death, dementia, or persistent physical disability, but there was a significantly higher risk of major hemorrhage in the aspirin group (3.8% vs 2.8%; hazard ratio, 1.38; 95% CI, 1.18-1.62; P < .001).
Several medical societies have updated their guideline recommendations for aspirin for primary prevention of CVD. The 2022 United States Public Service Task Force (USPSTF) provides a grade C recommendation (at least moderate certainty that the net benefit is small) to consider low-dose aspirin for the primary prevention of CVD on an individual patient basis for adults aged 40 to 59 years who have a ≥ 10% 10-year CVD risk. For adults aged ≥ 60 years, the USPSTF recommendation is grade D (moderate or high certainty that the practice has no net benefit or that harms outweigh the benefits) for low-dose aspirin use.1,3 The American College of Cardiology and American Heart Association (ACC/AHA) recommend considering low-dose aspirin for primary prevention of atherosclerotic cardiovascular disease (ASCVD) among select adults aged 40 to 70 years at higher CVD risk but not at increased risk of bleeding.4 The American Diabetes Association (ADA) recommends low-dose aspirin for primary prevention of CVD in patients with diabetes and additional risk factors such as family history of premature ASCVD, hypertension, dyslipidemia, smoking, or chronic kidney disease, and who are not at higher risk of bleeding.5 The ADA standards also caution against the use of aspirin as primary prevention in patients aged > 70 years. Low-dose aspirin use is not recommended for the primary prevention of CVD in older adults or adults of any age who are at increased risk of bleeding.
Recent literature using the US Department of Veterans Affairs (VA) Corporate Data Warehouse database confirms 86,555 of 1.8 million veterans aged > 70 years (5%) were taking low-dose aspirin for primary prevention of ASCVD despite guideline recommendations.6 Higher risk of gastrointestinal and other major bleeding from low-dose aspirin has been reported in the literature.1 Major bleeds represent a significant burden to the health care system with an estimated mean $13,093 cost for gastrointestinal bleed hospitalization.7
Considering the large scale aspirin use without appropriate indication within the veteran population, the risk of adverse effects, and the significant cost to patients and the health care system, it is imperative to determine the best approach to efficiently deprescribe aspirin for primary prevention among geriatric patients. Deprescribing refers to the systematic and supervised process of dose reduction or drug discontinuation with the goal of improving health and/or reducing the risk of adverse effects.8 During patient visits, primary care practitioners (PCPs) have opportunities to discontinue aspirin, but these encounters are time-limited and deprescribing might be secondary to more acute primary care needs. The shortage of PCPs is expected to worsen in coming years, which could further reduce their availability to assess inappropriate aspirin use.9
VA clinical pharmacist practitioners (CPPs) serve as medication experts and work autonomously under a broad scope of practice as part of the patient aligned care team.10-12 CPPs can free up time for PCPs and facilitate deprescribing efforts, especially for older adults. One retrospective cohort study conducted at a VA medical center found that CPPs deprescribed more potentially inappropriate medications among individuals aged ≥ 80 years compared with usual care with PCPs (26.8% vs 16.1%; P < .001).12,13 An aspirin deprescribing protocol conducted in 2022 resulted in nearly half of veterans aged ≥ 70 years contacted by phone agreeing to stop aspirin. Although this study supports the role pharmacists can play in reducing aspirin use in accordance with guidelines, the authors acknowledge that their interventions had a mean time of 12 minutes per patient and would require workflow changes.14 The purpose of this study is to evaluate the efficiency of aspirin deprescribing through 2 approaches: direct deprescribing by pharmacists using populationlevel review compared with clinicians following a pharmacist-led education.
Methods
This was a single-center quality improvement cohort study at the Durham VA Health Care System (DVAHCS) in North Carolina. Patients included were aged ≥ 70 years without known ASCVD who received care at any of 3 DVAHCS community-based outpatient clinics and prescribed aspirin. Patient data was obtained using the VIONE (Deprescribing Dashboard called Vital, Important, Optional, Not indicated, and Every medication has a specific indication or diagnosis) dashboard.15 VIONE was developed to identify potentially inappropriate medications (PIMs) that are eligible to deprescribe based on Beers Criteria, Screening Tool of Older Personsf Prescriptions criteria, and common clinical scenarios when clinicians determine the risk outweighs the benefit to continue a specific medication. 16,17 VIONE is used to reduce polypharmacy and improve patient safety, comfort, and medication adherence. Aspirin for patients aged ≥ 70 years without a history of ASCVD is a PIM identified by VIONE. Patients aged ≥ 70 years were chosen as an inclusion criteria in this study to match the ASPREE trial inclusion criteria and age inclusion criteria in the VIONE dashboard for aspirin deprescribing.2 Patient lists were generated for these potentially inappropriate aspirin prescriptions for 3 months before clinician staff education presentations, the day of the presentations, and 3 months after.
The primary endpoint was the number of veterans with aspirin deprescribed directly by 2 pharmacists over 12 weeks, divided by total patient care time spent, compared with the change in number of veterans with aspirin deprescribed by any DVAHCS physician, nurse practitioner, physician assistant, or CPP over 12 weeks, divided by the total pharmacist time spent on PCP education. Secondary endpoints were the number of aspirin orders discontinued by pharmacists and CPPs, the number of aspirin orders discontinued 12 weeks before pharmacist-led education compared with the number of aspirin orders discontinued 12 weeks after CPP-led education, average and median pharmacist time spent per patient encounter, and time of direct patient encounters vs time spent on PCP education.
Pharmacists reviewed each patient who met the inclusion criteria from the list generated by VIONE on December 1, 2022, for aspirin appropriateness according to the ACC/AHA and USPSTF guidelines, with the goal to discontinue aspirin for primary prevention of ASCVD and no other indications.1,4 Pharmacists documented their visits using VIONE methodology in the Computerized Patient Record System (CPRS) using a polypharmacy review note. CPPs contacted patients who were taking aspirin for primary prevention by unscheduled telephone call to assess for aspirin adherence, undocumented history of ASCVD, cardiovascular risk factors, and history of bleeding. Aspirin was discontinued if patients met guideline criteria recommendations and agreed to discontinuation. Risk-benefit discussions were completed when patients without known ASCVD were considered high risk because the ACC/AHA guidelines mention there is insufficient evidence of safety and efficacy of aspirin for primary prevention for patients with other known ASCVD risk factors (eg, strong family history of premature myocardial infarction, inability to achieve lipid, blood pressure, or glucose targets, or significant elevation in coronary artery calcium score).
High risk was defined as family history of premature ASCVD (in a male first-degree relative aged < 55 years or a female first-degree relative aged < 65 years), most recent blood pressure or 2 blood pressure results in the last 12 months > 160/100 mm Hg, recent hemoglobin A1c > 9%, and/or low-density lipoprotein > 190 mg/dL or not prescribed an indicated statin.3 Aspirin was continued or discontinued according to patient preference after the personalized risk-benefit discussion.
For patients with a clinical indication for aspirin use other than ASCVD (eg, atrial fibrillation not on anticoagulation, venous thromboembolism prophylaxis, carotid artery disease), CPPs documented their assessment and when appropriate deferred to the PCP for consideration of stopping aspirin. For patients with undocumented ASCVD, CPPs added their ASCVD history to their problem list and aspirin was continued. PCPs were notified by alert when aspirin was discontinued and when patients could not be reached by telephone.
presented a review of recent guideline updates and supporting literature at 2 online staff meetings. The education sessions lasted about 10 minutes and were presented to PCPs across 3 community-based outpatient clinics. An estimated 40 minutes were spent creating the PowerPoint education materials, seeking feedback, making edits, and answering questions or emails from PCPs after the presentation. During the presentation, pharmacists encouraged PCPs to discontinue aspirin (active VA prescriptions and reported over-the-counter use) for primary prevention of ASCVD in patients aged ≥ 70 years during their upcoming appointments and consider risk factors recommended by the ACC/AHA guidelines when applicable. PCPs were notified that CPPs planned to start a population review for discontinuing active VA aspirin prescriptions on December 1, 2022. The primary endpoint and secondary endpoints were analyzed using descriptive statistics. All data were analyzed using Microsoft Excel.

Results
A total of 868 patients aged ≥ 70 years with active prescriptions for aspirin were identified on December 1, 2022. After applying inclusion and exclusion criteria for the pharmacist population review, 224 patients were included for cohort final analysis (Figure). All 868 patients were eligible for the CPP intervention. Primary reasons for exclusion from the CPP population included over-thecounter aspirin and a history of ASCVD in the patient’s problem list. All patients were male, with a mean (SD) age of 75 (4.4) years (Table 1). Most patients were prescribed aspirin, 81 mg daily (n = 220; 98%).

The direct CPP deprescribing intervention resulted in 2 aspirin prescriptions discontinued per hour of pharmacist time and 67 aspirin prescriptions discontinued per hour of pharmacist time via the PCP education intervention. CPPs discontinued 66 aspirin orders in the 12 weeks before the PCP education sessions. A total of 230 aspirin prescriptions were discontinued in the 12 weeks following the PCP education sessions, with 97 discontinued directly by CPPs and 133 discontinued by PCPs. The PCP education session yielded an additional 67 discontinued aspirin orders compared with the 12 weeks before the education sessions (Table 2).

The CPP direct deprescribing intervention took about 48.3 hours, accounting for health record review and time interacting with patients. The PCP education intervention took about 60 minutes, which included time for preparing and delivering education materials (Table 3). CPP deprescribing encounter types, interventions, and related subcategories, and other identified indications to continue aspirin are listed in Table 4.


Discussion
Compared with direct deprescribing by pharmacists, the PCP education intervention was more efficient based on number of aspirin orders discontinued by pharmacist time. PCPs discontinued twice as many aspirin prescriptions in the 12 weeks after pharmacist-led education compared with the 12 weeks before.
Patients were primarily contacted by telephone (73%) for deprescribing. Among the 163 patients reached by phone and encouraged to discontinue aspirin, 97 patients (60%) accepted the recommendation, which was similar to the acceptance rates found in the literature (48% to 55%).14,18 Although many veterans continued taking aspirin (78%), most had indications for its continued use, such as a history of ASCVD, atrial fibrillation without anticoagulation, and carotid artery stenosis, and complex comorbidities that required further discussion with their PCP. Less common uses for aspirin were identified through CPRS review or patient reports included cerebral small vessel disease without history of ASCVD, subclavian artery stenosis, thrombocytosis, bioprosthetic valve replacement, giant cell arteritis, rheumatoid arthritis, and prevention of second eye involvement of ischemic optic neuropathy.
to describe the benefit of clinical pharmacy services for deprescribing aspirin for primary prevention of ASCVD through PCP education. Previously published literature has assessed alternative ways to identify or discontinue PIMs—including aspirin—among geriatric patients. One study evaluated the use of marking inappropriate aspirin prescriptions in the electronic health database, leading to a significant reduction in incidence of inappropriate aspirin prescribing; however, it did not assess changes in discontinuation rates of existing aspirin prescriptions.19 The previous VA pharmacist aspirin deprescribing protocol demonstrated pharmacists’ aptitude at discontinuing aspirin for primary prevention but only used direct patient contact and did not compare efficiency with other methods, including PCP education.14
This quality improvement project contributes new data to the existing literature to support the use of clinical pharmacists to discontinue aspirin for primary prevention and suggests a strong role for pharmacists as educators on clinical guidelines, in addition to their roles directly deprescribing PIMs in clinical practice. This study is further strengthened by its use of VIONE, which previously has demonstrated effectiveness in deprescribing a variety of PIMs in primary care settings.20
Despite using VIONE for generating a list of patients eligible for deprescription, our CPRS review found that this list was frequently inaccurate. For example, a small portion of patients were on the VIONE generated list indicating they had no ASCVD history, but had transient ischemic attack listed in their problem lists. Patient problem lists often were missing documented ASCVD history that was revealed by patient interview or CPRS review. It is possible that patients interviewed might have omitted relevant ASCVD history because of low health literacy, conditions affecting memory, or use of health care services outside the VA system.
There were several instances of aspirin used for other non-ASCVD indications, such as primary stroke prevention in atrial fibrillation. The ACC/AHA atrial fibrillation guidelines previously provided a Class IIb recommendation (benefit is greater than risk but additional studies are needed) for considering no antithrombic therapy or treatment with oral anticoagulant or aspirin for nonvalvular atrial fibrillation with CHA2DS2-VASc (Congestive heart failure, Hypertension, Age [> 65 y, 1 point; > 75 y, 2 points], Diabetes, previous Stroke/transient ischemic attack [2 points]) score of 1.21 The ACC/ AHA guidelines were updated in 2023 to recommend against antiplatelet therapy as an alternative to anticoagulation for reducing cardioembolic stroke risk among patients with atrial fibrillation with no indication for antiplatelet therapy because of risk of harm.22 If a patient has no risk factors for stroke, aspirin is not recommended to prevent thromboembolic events because of a lack of benefit. Interventions from this quality improvement study were completed before the 2023 atrial fibrillation guideline was published and therefore in this study aspirin was not discontinued when used for atrial fibrillation. Aspirin use for atrial fibrillation might benefit from similar discontinuation efforts analyzed within this study. Beyond atrial fibrillation, major guidelines do not comment on the use of aspirin for any other indications in the absence of clinical ASCVD.
Limitations
This study is limited by the lack of clinical consensus for complex patients and demonstrates the importance of individualized patient assessment when considering discontinuing aspirin. Because of the project’s relatively short intervention period, aspirin deprescribing rates could decrease over time and repeated education efforts might be necessary to see lasting impact. Health care professionals from services outside of primary care also might have discontinued aspirin during the study period unrelated to the education and these discontinued aspirin prescriptions could contribute to the higher rate observed among PCPs. This study included a specific population cohort of male, US veterans and might not reflect other populations where these interventions could be implemented.
The measurement of time spent by pharmacists and PCPs is an additional limitation. Although it is expected that PCPs attempt to discontinue aspirin during their existing patient care appointments, the time spent during visits was not measured or documented. Direct deprescribing by pharmacist CPRS review required a significant amount of time and could be a barrier to successful intervention by CPPs in patient aligned care teams.
To reduce the time pharmacists spent completing CPRS reviews, an aspirin deprescribing clinical reminder tool could be used to assess use and appropriate indication quickly during any primary care visit led by a PCP or CPP. In addition, it is recommended that pharmacists regularly educate health care professionals on guideline recommendations for aspirin use among geriatric patients. Future studies of the incidence of major cardiovascular events after aspirin deprescribing among geriatric patients and a longitudinal cost/benefit analysis could support these initiatives.
Conclusions
In this study, pharmacists successfully deprescribed inappropriate medications, such as aspirin. However, pharmacist-led PCP education is more efficient compared with direct deprescribing using a population-level review. PCP education requires less time and could allow ambulatory care pharmacists to spend more time on other direct patient care interventions to improve quality and access to care in primary care clinics. This study’s results further support the role of pharmacists in deprescribing PIMs for older adults and the use of a deprescribing tool, such as VIONE, in a primary care setting.
- US Preventive Services Task Force; Davidson KW, Barry MJ, et al. Aspirin use to prevent cardiovascular disease: US Preventive Services Task Force recommendation statement. JAMA. 2022;327(16):1577-1584. doi:10.1001/jama.2022.4983
- McNeil JJ, Nelson MR, Woods RL, et al. Effect of aspirin on all-cause mortality in the healthy elderly. N Engl J Med. 2018;379(16):1519-1528. doi:10.1056/NEJMoa1803955
- Barry MJ, Wolff TA, Pbert L, et al. Putting evidence into practice: an update on the US Preventive Services Task Force methods for developing recommendations for preventive services. Ann Fam Med. 2023;21(2):165-171. doi:10.1370/afm.2946
- Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/ AHA Guideline on the Primary Prevention of Cardiovascular Disease: A report of the American College of Cardiology/ American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;140(11):e596-e646. doi:10.1161/CIR.0000000000000678
- American Diabetes Association Professional Practice Committee. 10. Cardiovascular disease and risk management: Standards of care in diabetes-2024. Diabetes Care. 2024;47(Suppl 1):S179-S218. doi:10.2337/dc24-S010
- Ong SY, Chui P, Bhargava A, Justice A, Hauser RG. Estimating aspirin overuse for primary prevention of atherosclerotic cardiovascular disease (from a nationwide healthcare system). Am J Cardiol. 2020;137:25-30. doi:10.1016/j.amjcard.2020.09.042
- Weiss AJ, Jiang HJ. Overview of clinical conditions with frequent and costly hospital readmissions by payer, 2018. In: Healthcare Cost and Utilization Project (HCUP) Statistical Briefs. Agency for Healthcare Research and Quality (US); July 20, 2021.
- Krishnaswami A, Steinman MA, Goyal P, et al. Deprescribing in older adults with cardiovascular disease. J Am Coll Cardiol. 2019;73(20):2584-2595. doi:10.1016/j.jacc.2019.03.467
- Association of American Medical Colleges. The complexities of physician supply and demand: projections from 2019 to 2034. Accessed March 17, 2024. https://www.aamc.org/media/54681/download
- US Department of Veterans Affairs, Veterans Health Administration. VHA Directive 1108.07(1): General pharmacy service requirements. November 28, 2022. Accessed March 17, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=10045
- US Department of Veterans Affairs, Veterans Health Administration. VHA Handbook 1108.11(3): Clinical pharmacy services. July 1, 2015. Accessed March 17, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=3120
- US Department of Veterans Affairs. Clinical pharmacist practitioner (CPP) to improve access to and quality of care August 2021. August 2021. Accessed May 19, 2023. https://www.pbm.va.gov/PBM/CPPO/Documents/ExternalFactSheet_OptimizingtheCPPToImproveAccess_508.pdf
- Ammerman CA, Simpkins BA, Warman N, Downs TN. Potentially inappropriate medications in older adults: Deprescribing with a clinical pharmacist. J Am Geriatr Soc. 2019;67(1):115-118. doi:10.1111/jgs.15623
- Rothbauer K, Siodlak M, Dreischmeier E, Ranola TS, Welch L. Evaluation of a pharmacist-driven ambulatory aspirin deprescribing protocol. Fed Pract. 2022;39(suppl 5):S37- S41a. doi:10.12788/fp.0294
- US Department of Veterans Affairs. VIONE changes the way VA handles prescriptions. January 25, 2020. Accessed May 21, 2023. https://news.va.gov/70709/vione-changes-way-va-handles-prescriptions/
- 2023 American Geriatrics Society Beers Criteria Update Expert Panel. American Geriatrics Society 2023 updated AGS Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2023;71(7):2052- 2081. doi:10.1111/jgs.18372
- O’Mahony D, Cherubini A, Guiteras AR, et al. STOPP/ START criteria for potentially inappropriate prescribing in older people: version 3. Eur Geriatr Med. 2023;14(4):625- 632. doi:10.1007/s41999-023-00777-y
- Draeger C, Lodhi F, Geissinger N, Larson T, Griesbach S. Interdisciplinary deprescribing of aspirin through prescriber education and provision of patient-specific recommendations. WMJ. 2022;121(3):220-225
- de Lusignan S, Hinton W, Seidu S, et al. Dashboards to reduce inappropriate prescribing of metformin and aspirin: A quality assurance programme in a primary care sentinel network. Prim Care Diabetes. 2021;15(6):1075-1079. doi:10.1016/j.pcd.2021.06.003
- Nelson MW, Downs TN, Puglisi GM, Simpkins BA, Collier AS. Use of a deprescribing tool in an interdisciplinary primary-care patient-aligned care team. Sr Care Pharm. 2022;37(1):34-43. doi:10.4140/TCP.n.2022.34
- January CT, Wann LS, Alpert JS, et al. 2014 AHA/ ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the Heart Rhythm Society. Circulation. 2014;130(23):e199-e267. doi:10.1161/CIR.0000000000000041
- Joglar JA, Chung MK, Armbruster AL, et al. 2023 ACC/ AHA/ACCP/HRS Guideline for the Diagnosis and Management of Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines Circulation. 2024;149(1):e1- e156. doi:10.1161/CIR.0000000000001193
Low-dose aspirin commonly is used for the prevention of cardiovascular disease (CVD) but is associated with an increased risk of major bleeding.1 The use of aspirin for primary prevention is largely extrapolated from clinical trials showing benefit in the secondary prevention of myocardial infarction and ischemic stroke. However, results from the Aspirin in Reducing Events in the Elderly (ASPREE) trial challenged this practice.2 The ASPREE trial, conducted in the United States and Australia from 2010 to 2014, sought to determine whether daily 100 mg aspirin, was superior to placebo in promoting disability-free survival among older adults. Participants were aged ≥ 70 years (≥ 65 years for Hispanic and Black US participants), living in the community, and were free from preexisting CVD, cerebrovascular disease, or any chronic condition likely to limit survival to < 5 years. The study found no significant difference in the primary endpoints of death, dementia, or persistent physical disability, but there was a significantly higher risk of major hemorrhage in the aspirin group (3.8% vs 2.8%; hazard ratio, 1.38; 95% CI, 1.18-1.62; P < .001).
Several medical societies have updated their guideline recommendations for aspirin for primary prevention of CVD. The 2022 United States Public Service Task Force (USPSTF) provides a grade C recommendation (at least moderate certainty that the net benefit is small) to consider low-dose aspirin for the primary prevention of CVD on an individual patient basis for adults aged 40 to 59 years who have a ≥ 10% 10-year CVD risk. For adults aged ≥ 60 years, the USPSTF recommendation is grade D (moderate or high certainty that the practice has no net benefit or that harms outweigh the benefits) for low-dose aspirin use.1,3 The American College of Cardiology and American Heart Association (ACC/AHA) recommend considering low-dose aspirin for primary prevention of atherosclerotic cardiovascular disease (ASCVD) among select adults aged 40 to 70 years at higher CVD risk but not at increased risk of bleeding.4 The American Diabetes Association (ADA) recommends low-dose aspirin for primary prevention of CVD in patients with diabetes and additional risk factors such as family history of premature ASCVD, hypertension, dyslipidemia, smoking, or chronic kidney disease, and who are not at higher risk of bleeding.5 The ADA standards also caution against the use of aspirin as primary prevention in patients aged > 70 years. Low-dose aspirin use is not recommended for the primary prevention of CVD in older adults or adults of any age who are at increased risk of bleeding.
Recent literature using the US Department of Veterans Affairs (VA) Corporate Data Warehouse database confirms 86,555 of 1.8 million veterans aged > 70 years (5%) were taking low-dose aspirin for primary prevention of ASCVD despite guideline recommendations.6 Higher risk of gastrointestinal and other major bleeding from low-dose aspirin has been reported in the literature.1 Major bleeds represent a significant burden to the health care system with an estimated mean $13,093 cost for gastrointestinal bleed hospitalization.7
Considering the large scale aspirin use without appropriate indication within the veteran population, the risk of adverse effects, and the significant cost to patients and the health care system, it is imperative to determine the best approach to efficiently deprescribe aspirin for primary prevention among geriatric patients. Deprescribing refers to the systematic and supervised process of dose reduction or drug discontinuation with the goal of improving health and/or reducing the risk of adverse effects.8 During patient visits, primary care practitioners (PCPs) have opportunities to discontinue aspirin, but these encounters are time-limited and deprescribing might be secondary to more acute primary care needs. The shortage of PCPs is expected to worsen in coming years, which could further reduce their availability to assess inappropriate aspirin use.9
VA clinical pharmacist practitioners (CPPs) serve as medication experts and work autonomously under a broad scope of practice as part of the patient aligned care team.10-12 CPPs can free up time for PCPs and facilitate deprescribing efforts, especially for older adults. One retrospective cohort study conducted at a VA medical center found that CPPs deprescribed more potentially inappropriate medications among individuals aged ≥ 80 years compared with usual care with PCPs (26.8% vs 16.1%; P < .001).12,13 An aspirin deprescribing protocol conducted in 2022 resulted in nearly half of veterans aged ≥ 70 years contacted by phone agreeing to stop aspirin. Although this study supports the role pharmacists can play in reducing aspirin use in accordance with guidelines, the authors acknowledge that their interventions had a mean time of 12 minutes per patient and would require workflow changes.14 The purpose of this study is to evaluate the efficiency of aspirin deprescribing through 2 approaches: direct deprescribing by pharmacists using populationlevel review compared with clinicians following a pharmacist-led education.
Methods
This was a single-center quality improvement cohort study at the Durham VA Health Care System (DVAHCS) in North Carolina. Patients included were aged ≥ 70 years without known ASCVD who received care at any of 3 DVAHCS community-based outpatient clinics and prescribed aspirin. Patient data was obtained using the VIONE (Deprescribing Dashboard called Vital, Important, Optional, Not indicated, and Every medication has a specific indication or diagnosis) dashboard.15 VIONE was developed to identify potentially inappropriate medications (PIMs) that are eligible to deprescribe based on Beers Criteria, Screening Tool of Older Personsf Prescriptions criteria, and common clinical scenarios when clinicians determine the risk outweighs the benefit to continue a specific medication. 16,17 VIONE is used to reduce polypharmacy and improve patient safety, comfort, and medication adherence. Aspirin for patients aged ≥ 70 years without a history of ASCVD is a PIM identified by VIONE. Patients aged ≥ 70 years were chosen as an inclusion criteria in this study to match the ASPREE trial inclusion criteria and age inclusion criteria in the VIONE dashboard for aspirin deprescribing.2 Patient lists were generated for these potentially inappropriate aspirin prescriptions for 3 months before clinician staff education presentations, the day of the presentations, and 3 months after.
The primary endpoint was the number of veterans with aspirin deprescribed directly by 2 pharmacists over 12 weeks, divided by total patient care time spent, compared with the change in number of veterans with aspirin deprescribed by any DVAHCS physician, nurse practitioner, physician assistant, or CPP over 12 weeks, divided by the total pharmacist time spent on PCP education. Secondary endpoints were the number of aspirin orders discontinued by pharmacists and CPPs, the number of aspirin orders discontinued 12 weeks before pharmacist-led education compared with the number of aspirin orders discontinued 12 weeks after CPP-led education, average and median pharmacist time spent per patient encounter, and time of direct patient encounters vs time spent on PCP education.
Pharmacists reviewed each patient who met the inclusion criteria from the list generated by VIONE on December 1, 2022, for aspirin appropriateness according to the ACC/AHA and USPSTF guidelines, with the goal to discontinue aspirin for primary prevention of ASCVD and no other indications.1,4 Pharmacists documented their visits using VIONE methodology in the Computerized Patient Record System (CPRS) using a polypharmacy review note. CPPs contacted patients who were taking aspirin for primary prevention by unscheduled telephone call to assess for aspirin adherence, undocumented history of ASCVD, cardiovascular risk factors, and history of bleeding. Aspirin was discontinued if patients met guideline criteria recommendations and agreed to discontinuation. Risk-benefit discussions were completed when patients without known ASCVD were considered high risk because the ACC/AHA guidelines mention there is insufficient evidence of safety and efficacy of aspirin for primary prevention for patients with other known ASCVD risk factors (eg, strong family history of premature myocardial infarction, inability to achieve lipid, blood pressure, or glucose targets, or significant elevation in coronary artery calcium score).
High risk was defined as family history of premature ASCVD (in a male first-degree relative aged < 55 years or a female first-degree relative aged < 65 years), most recent blood pressure or 2 blood pressure results in the last 12 months > 160/100 mm Hg, recent hemoglobin A1c > 9%, and/or low-density lipoprotein > 190 mg/dL or not prescribed an indicated statin.3 Aspirin was continued or discontinued according to patient preference after the personalized risk-benefit discussion.
For patients with a clinical indication for aspirin use other than ASCVD (eg, atrial fibrillation not on anticoagulation, venous thromboembolism prophylaxis, carotid artery disease), CPPs documented their assessment and when appropriate deferred to the PCP for consideration of stopping aspirin. For patients with undocumented ASCVD, CPPs added their ASCVD history to their problem list and aspirin was continued. PCPs were notified by alert when aspirin was discontinued and when patients could not be reached by telephone.
presented a review of recent guideline updates and supporting literature at 2 online staff meetings. The education sessions lasted about 10 minutes and were presented to PCPs across 3 community-based outpatient clinics. An estimated 40 minutes were spent creating the PowerPoint education materials, seeking feedback, making edits, and answering questions or emails from PCPs after the presentation. During the presentation, pharmacists encouraged PCPs to discontinue aspirin (active VA prescriptions and reported over-the-counter use) for primary prevention of ASCVD in patients aged ≥ 70 years during their upcoming appointments and consider risk factors recommended by the ACC/AHA guidelines when applicable. PCPs were notified that CPPs planned to start a population review for discontinuing active VA aspirin prescriptions on December 1, 2022. The primary endpoint and secondary endpoints were analyzed using descriptive statistics. All data were analyzed using Microsoft Excel.

Results
A total of 868 patients aged ≥ 70 years with active prescriptions for aspirin were identified on December 1, 2022. After applying inclusion and exclusion criteria for the pharmacist population review, 224 patients were included for cohort final analysis (Figure). All 868 patients were eligible for the CPP intervention. Primary reasons for exclusion from the CPP population included over-thecounter aspirin and a history of ASCVD in the patient’s problem list. All patients were male, with a mean (SD) age of 75 (4.4) years (Table 1). Most patients were prescribed aspirin, 81 mg daily (n = 220; 98%).

The direct CPP deprescribing intervention resulted in 2 aspirin prescriptions discontinued per hour of pharmacist time and 67 aspirin prescriptions discontinued per hour of pharmacist time via the PCP education intervention. CPPs discontinued 66 aspirin orders in the 12 weeks before the PCP education sessions. A total of 230 aspirin prescriptions were discontinued in the 12 weeks following the PCP education sessions, with 97 discontinued directly by CPPs and 133 discontinued by PCPs. The PCP education session yielded an additional 67 discontinued aspirin orders compared with the 12 weeks before the education sessions (Table 2).

The CPP direct deprescribing intervention took about 48.3 hours, accounting for health record review and time interacting with patients. The PCP education intervention took about 60 minutes, which included time for preparing and delivering education materials (Table 3). CPP deprescribing encounter types, interventions, and related subcategories, and other identified indications to continue aspirin are listed in Table 4.


Discussion
Compared with direct deprescribing by pharmacists, the PCP education intervention was more efficient based on number of aspirin orders discontinued by pharmacist time. PCPs discontinued twice as many aspirin prescriptions in the 12 weeks after pharmacist-led education compared with the 12 weeks before.
Patients were primarily contacted by telephone (73%) for deprescribing. Among the 163 patients reached by phone and encouraged to discontinue aspirin, 97 patients (60%) accepted the recommendation, which was similar to the acceptance rates found in the literature (48% to 55%).14,18 Although many veterans continued taking aspirin (78%), most had indications for its continued use, such as a history of ASCVD, atrial fibrillation without anticoagulation, and carotid artery stenosis, and complex comorbidities that required further discussion with their PCP. Less common uses for aspirin were identified through CPRS review or patient reports included cerebral small vessel disease without history of ASCVD, subclavian artery stenosis, thrombocytosis, bioprosthetic valve replacement, giant cell arteritis, rheumatoid arthritis, and prevention of second eye involvement of ischemic optic neuropathy.
to describe the benefit of clinical pharmacy services for deprescribing aspirin for primary prevention of ASCVD through PCP education. Previously published literature has assessed alternative ways to identify or discontinue PIMs—including aspirin—among geriatric patients. One study evaluated the use of marking inappropriate aspirin prescriptions in the electronic health database, leading to a significant reduction in incidence of inappropriate aspirin prescribing; however, it did not assess changes in discontinuation rates of existing aspirin prescriptions.19 The previous VA pharmacist aspirin deprescribing protocol demonstrated pharmacists’ aptitude at discontinuing aspirin for primary prevention but only used direct patient contact and did not compare efficiency with other methods, including PCP education.14
This quality improvement project contributes new data to the existing literature to support the use of clinical pharmacists to discontinue aspirin for primary prevention and suggests a strong role for pharmacists as educators on clinical guidelines, in addition to their roles directly deprescribing PIMs in clinical practice. This study is further strengthened by its use of VIONE, which previously has demonstrated effectiveness in deprescribing a variety of PIMs in primary care settings.20
Despite using VIONE for generating a list of patients eligible for deprescription, our CPRS review found that this list was frequently inaccurate. For example, a small portion of patients were on the VIONE generated list indicating they had no ASCVD history, but had transient ischemic attack listed in their problem lists. Patient problem lists often were missing documented ASCVD history that was revealed by patient interview or CPRS review. It is possible that patients interviewed might have omitted relevant ASCVD history because of low health literacy, conditions affecting memory, or use of health care services outside the VA system.
There were several instances of aspirin used for other non-ASCVD indications, such as primary stroke prevention in atrial fibrillation. The ACC/AHA atrial fibrillation guidelines previously provided a Class IIb recommendation (benefit is greater than risk but additional studies are needed) for considering no antithrombic therapy or treatment with oral anticoagulant or aspirin for nonvalvular atrial fibrillation with CHA2DS2-VASc (Congestive heart failure, Hypertension, Age [> 65 y, 1 point; > 75 y, 2 points], Diabetes, previous Stroke/transient ischemic attack [2 points]) score of 1.21 The ACC/ AHA guidelines were updated in 2023 to recommend against antiplatelet therapy as an alternative to anticoagulation for reducing cardioembolic stroke risk among patients with atrial fibrillation with no indication for antiplatelet therapy because of risk of harm.22 If a patient has no risk factors for stroke, aspirin is not recommended to prevent thromboembolic events because of a lack of benefit. Interventions from this quality improvement study were completed before the 2023 atrial fibrillation guideline was published and therefore in this study aspirin was not discontinued when used for atrial fibrillation. Aspirin use for atrial fibrillation might benefit from similar discontinuation efforts analyzed within this study. Beyond atrial fibrillation, major guidelines do not comment on the use of aspirin for any other indications in the absence of clinical ASCVD.
Limitations
This study is limited by the lack of clinical consensus for complex patients and demonstrates the importance of individualized patient assessment when considering discontinuing aspirin. Because of the project’s relatively short intervention period, aspirin deprescribing rates could decrease over time and repeated education efforts might be necessary to see lasting impact. Health care professionals from services outside of primary care also might have discontinued aspirin during the study period unrelated to the education and these discontinued aspirin prescriptions could contribute to the higher rate observed among PCPs. This study included a specific population cohort of male, US veterans and might not reflect other populations where these interventions could be implemented.
The measurement of time spent by pharmacists and PCPs is an additional limitation. Although it is expected that PCPs attempt to discontinue aspirin during their existing patient care appointments, the time spent during visits was not measured or documented. Direct deprescribing by pharmacist CPRS review required a significant amount of time and could be a barrier to successful intervention by CPPs in patient aligned care teams.
To reduce the time pharmacists spent completing CPRS reviews, an aspirin deprescribing clinical reminder tool could be used to assess use and appropriate indication quickly during any primary care visit led by a PCP or CPP. In addition, it is recommended that pharmacists regularly educate health care professionals on guideline recommendations for aspirin use among geriatric patients. Future studies of the incidence of major cardiovascular events after aspirin deprescribing among geriatric patients and a longitudinal cost/benefit analysis could support these initiatives.
Conclusions
In this study, pharmacists successfully deprescribed inappropriate medications, such as aspirin. However, pharmacist-led PCP education is more efficient compared with direct deprescribing using a population-level review. PCP education requires less time and could allow ambulatory care pharmacists to spend more time on other direct patient care interventions to improve quality and access to care in primary care clinics. This study’s results further support the role of pharmacists in deprescribing PIMs for older adults and the use of a deprescribing tool, such as VIONE, in a primary care setting.
Low-dose aspirin commonly is used for the prevention of cardiovascular disease (CVD) but is associated with an increased risk of major bleeding.1 The use of aspirin for primary prevention is largely extrapolated from clinical trials showing benefit in the secondary prevention of myocardial infarction and ischemic stroke. However, results from the Aspirin in Reducing Events in the Elderly (ASPREE) trial challenged this practice.2 The ASPREE trial, conducted in the United States and Australia from 2010 to 2014, sought to determine whether daily 100 mg aspirin, was superior to placebo in promoting disability-free survival among older adults. Participants were aged ≥ 70 years (≥ 65 years for Hispanic and Black US participants), living in the community, and were free from preexisting CVD, cerebrovascular disease, or any chronic condition likely to limit survival to < 5 years. The study found no significant difference in the primary endpoints of death, dementia, or persistent physical disability, but there was a significantly higher risk of major hemorrhage in the aspirin group (3.8% vs 2.8%; hazard ratio, 1.38; 95% CI, 1.18-1.62; P < .001).
Several medical societies have updated their guideline recommendations for aspirin for primary prevention of CVD. The 2022 United States Public Service Task Force (USPSTF) provides a grade C recommendation (at least moderate certainty that the net benefit is small) to consider low-dose aspirin for the primary prevention of CVD on an individual patient basis for adults aged 40 to 59 years who have a ≥ 10% 10-year CVD risk. For adults aged ≥ 60 years, the USPSTF recommendation is grade D (moderate or high certainty that the practice has no net benefit or that harms outweigh the benefits) for low-dose aspirin use.1,3 The American College of Cardiology and American Heart Association (ACC/AHA) recommend considering low-dose aspirin for primary prevention of atherosclerotic cardiovascular disease (ASCVD) among select adults aged 40 to 70 years at higher CVD risk but not at increased risk of bleeding.4 The American Diabetes Association (ADA) recommends low-dose aspirin for primary prevention of CVD in patients with diabetes and additional risk factors such as family history of premature ASCVD, hypertension, dyslipidemia, smoking, or chronic kidney disease, and who are not at higher risk of bleeding.5 The ADA standards also caution against the use of aspirin as primary prevention in patients aged > 70 years. Low-dose aspirin use is not recommended for the primary prevention of CVD in older adults or adults of any age who are at increased risk of bleeding.
Recent literature using the US Department of Veterans Affairs (VA) Corporate Data Warehouse database confirms 86,555 of 1.8 million veterans aged > 70 years (5%) were taking low-dose aspirin for primary prevention of ASCVD despite guideline recommendations.6 Higher risk of gastrointestinal and other major bleeding from low-dose aspirin has been reported in the literature.1 Major bleeds represent a significant burden to the health care system with an estimated mean $13,093 cost for gastrointestinal bleed hospitalization.7
Considering the large scale aspirin use without appropriate indication within the veteran population, the risk of adverse effects, and the significant cost to patients and the health care system, it is imperative to determine the best approach to efficiently deprescribe aspirin for primary prevention among geriatric patients. Deprescribing refers to the systematic and supervised process of dose reduction or drug discontinuation with the goal of improving health and/or reducing the risk of adverse effects.8 During patient visits, primary care practitioners (PCPs) have opportunities to discontinue aspirin, but these encounters are time-limited and deprescribing might be secondary to more acute primary care needs. The shortage of PCPs is expected to worsen in coming years, which could further reduce their availability to assess inappropriate aspirin use.9
VA clinical pharmacist practitioners (CPPs) serve as medication experts and work autonomously under a broad scope of practice as part of the patient aligned care team.10-12 CPPs can free up time for PCPs and facilitate deprescribing efforts, especially for older adults. One retrospective cohort study conducted at a VA medical center found that CPPs deprescribed more potentially inappropriate medications among individuals aged ≥ 80 years compared with usual care with PCPs (26.8% vs 16.1%; P < .001).12,13 An aspirin deprescribing protocol conducted in 2022 resulted in nearly half of veterans aged ≥ 70 years contacted by phone agreeing to stop aspirin. Although this study supports the role pharmacists can play in reducing aspirin use in accordance with guidelines, the authors acknowledge that their interventions had a mean time of 12 minutes per patient and would require workflow changes.14 The purpose of this study is to evaluate the efficiency of aspirin deprescribing through 2 approaches: direct deprescribing by pharmacists using populationlevel review compared with clinicians following a pharmacist-led education.
Methods
This was a single-center quality improvement cohort study at the Durham VA Health Care System (DVAHCS) in North Carolina. Patients included were aged ≥ 70 years without known ASCVD who received care at any of 3 DVAHCS community-based outpatient clinics and prescribed aspirin. Patient data was obtained using the VIONE (Deprescribing Dashboard called Vital, Important, Optional, Not indicated, and Every medication has a specific indication or diagnosis) dashboard.15 VIONE was developed to identify potentially inappropriate medications (PIMs) that are eligible to deprescribe based on Beers Criteria, Screening Tool of Older Personsf Prescriptions criteria, and common clinical scenarios when clinicians determine the risk outweighs the benefit to continue a specific medication. 16,17 VIONE is used to reduce polypharmacy and improve patient safety, comfort, and medication adherence. Aspirin for patients aged ≥ 70 years without a history of ASCVD is a PIM identified by VIONE. Patients aged ≥ 70 years were chosen as an inclusion criteria in this study to match the ASPREE trial inclusion criteria and age inclusion criteria in the VIONE dashboard for aspirin deprescribing.2 Patient lists were generated for these potentially inappropriate aspirin prescriptions for 3 months before clinician staff education presentations, the day of the presentations, and 3 months after.
The primary endpoint was the number of veterans with aspirin deprescribed directly by 2 pharmacists over 12 weeks, divided by total patient care time spent, compared with the change in number of veterans with aspirin deprescribed by any DVAHCS physician, nurse practitioner, physician assistant, or CPP over 12 weeks, divided by the total pharmacist time spent on PCP education. Secondary endpoints were the number of aspirin orders discontinued by pharmacists and CPPs, the number of aspirin orders discontinued 12 weeks before pharmacist-led education compared with the number of aspirin orders discontinued 12 weeks after CPP-led education, average and median pharmacist time spent per patient encounter, and time of direct patient encounters vs time spent on PCP education.
Pharmacists reviewed each patient who met the inclusion criteria from the list generated by VIONE on December 1, 2022, for aspirin appropriateness according to the ACC/AHA and USPSTF guidelines, with the goal to discontinue aspirin for primary prevention of ASCVD and no other indications.1,4 Pharmacists documented their visits using VIONE methodology in the Computerized Patient Record System (CPRS) using a polypharmacy review note. CPPs contacted patients who were taking aspirin for primary prevention by unscheduled telephone call to assess for aspirin adherence, undocumented history of ASCVD, cardiovascular risk factors, and history of bleeding. Aspirin was discontinued if patients met guideline criteria recommendations and agreed to discontinuation. Risk-benefit discussions were completed when patients without known ASCVD were considered high risk because the ACC/AHA guidelines mention there is insufficient evidence of safety and efficacy of aspirin for primary prevention for patients with other known ASCVD risk factors (eg, strong family history of premature myocardial infarction, inability to achieve lipid, blood pressure, or glucose targets, or significant elevation in coronary artery calcium score).
High risk was defined as family history of premature ASCVD (in a male first-degree relative aged < 55 years or a female first-degree relative aged < 65 years), most recent blood pressure or 2 blood pressure results in the last 12 months > 160/100 mm Hg, recent hemoglobin A1c > 9%, and/or low-density lipoprotein > 190 mg/dL or not prescribed an indicated statin.3 Aspirin was continued or discontinued according to patient preference after the personalized risk-benefit discussion.
For patients with a clinical indication for aspirin use other than ASCVD (eg, atrial fibrillation not on anticoagulation, venous thromboembolism prophylaxis, carotid artery disease), CPPs documented their assessment and when appropriate deferred to the PCP for consideration of stopping aspirin. For patients with undocumented ASCVD, CPPs added their ASCVD history to their problem list and aspirin was continued. PCPs were notified by alert when aspirin was discontinued and when patients could not be reached by telephone.
presented a review of recent guideline updates and supporting literature at 2 online staff meetings. The education sessions lasted about 10 minutes and were presented to PCPs across 3 community-based outpatient clinics. An estimated 40 minutes were spent creating the PowerPoint education materials, seeking feedback, making edits, and answering questions or emails from PCPs after the presentation. During the presentation, pharmacists encouraged PCPs to discontinue aspirin (active VA prescriptions and reported over-the-counter use) for primary prevention of ASCVD in patients aged ≥ 70 years during their upcoming appointments and consider risk factors recommended by the ACC/AHA guidelines when applicable. PCPs were notified that CPPs planned to start a population review for discontinuing active VA aspirin prescriptions on December 1, 2022. The primary endpoint and secondary endpoints were analyzed using descriptive statistics. All data were analyzed using Microsoft Excel.

Results
A total of 868 patients aged ≥ 70 years with active prescriptions for aspirin were identified on December 1, 2022. After applying inclusion and exclusion criteria for the pharmacist population review, 224 patients were included for cohort final analysis (Figure). All 868 patients were eligible for the CPP intervention. Primary reasons for exclusion from the CPP population included over-thecounter aspirin and a history of ASCVD in the patient’s problem list. All patients were male, with a mean (SD) age of 75 (4.4) years (Table 1). Most patients were prescribed aspirin, 81 mg daily (n = 220; 98%).

The direct CPP deprescribing intervention resulted in 2 aspirin prescriptions discontinued per hour of pharmacist time and 67 aspirin prescriptions discontinued per hour of pharmacist time via the PCP education intervention. CPPs discontinued 66 aspirin orders in the 12 weeks before the PCP education sessions. A total of 230 aspirin prescriptions were discontinued in the 12 weeks following the PCP education sessions, with 97 discontinued directly by CPPs and 133 discontinued by PCPs. The PCP education session yielded an additional 67 discontinued aspirin orders compared with the 12 weeks before the education sessions (Table 2).

The CPP direct deprescribing intervention took about 48.3 hours, accounting for health record review and time interacting with patients. The PCP education intervention took about 60 minutes, which included time for preparing and delivering education materials (Table 3). CPP deprescribing encounter types, interventions, and related subcategories, and other identified indications to continue aspirin are listed in Table 4.


Discussion
Compared with direct deprescribing by pharmacists, the PCP education intervention was more efficient based on number of aspirin orders discontinued by pharmacist time. PCPs discontinued twice as many aspirin prescriptions in the 12 weeks after pharmacist-led education compared with the 12 weeks before.
Patients were primarily contacted by telephone (73%) for deprescribing. Among the 163 patients reached by phone and encouraged to discontinue aspirin, 97 patients (60%) accepted the recommendation, which was similar to the acceptance rates found in the literature (48% to 55%).14,18 Although many veterans continued taking aspirin (78%), most had indications for its continued use, such as a history of ASCVD, atrial fibrillation without anticoagulation, and carotid artery stenosis, and complex comorbidities that required further discussion with their PCP. Less common uses for aspirin were identified through CPRS review or patient reports included cerebral small vessel disease without history of ASCVD, subclavian artery stenosis, thrombocytosis, bioprosthetic valve replacement, giant cell arteritis, rheumatoid arthritis, and prevention of second eye involvement of ischemic optic neuropathy.
to describe the benefit of clinical pharmacy services for deprescribing aspirin for primary prevention of ASCVD through PCP education. Previously published literature has assessed alternative ways to identify or discontinue PIMs—including aspirin—among geriatric patients. One study evaluated the use of marking inappropriate aspirin prescriptions in the electronic health database, leading to a significant reduction in incidence of inappropriate aspirin prescribing; however, it did not assess changes in discontinuation rates of existing aspirin prescriptions.19 The previous VA pharmacist aspirin deprescribing protocol demonstrated pharmacists’ aptitude at discontinuing aspirin for primary prevention but only used direct patient contact and did not compare efficiency with other methods, including PCP education.14
This quality improvement project contributes new data to the existing literature to support the use of clinical pharmacists to discontinue aspirin for primary prevention and suggests a strong role for pharmacists as educators on clinical guidelines, in addition to their roles directly deprescribing PIMs in clinical practice. This study is further strengthened by its use of VIONE, which previously has demonstrated effectiveness in deprescribing a variety of PIMs in primary care settings.20
Despite using VIONE for generating a list of patients eligible for deprescription, our CPRS review found that this list was frequently inaccurate. For example, a small portion of patients were on the VIONE generated list indicating they had no ASCVD history, but had transient ischemic attack listed in their problem lists. Patient problem lists often were missing documented ASCVD history that was revealed by patient interview or CPRS review. It is possible that patients interviewed might have omitted relevant ASCVD history because of low health literacy, conditions affecting memory, or use of health care services outside the VA system.
There were several instances of aspirin used for other non-ASCVD indications, such as primary stroke prevention in atrial fibrillation. The ACC/AHA atrial fibrillation guidelines previously provided a Class IIb recommendation (benefit is greater than risk but additional studies are needed) for considering no antithrombic therapy or treatment with oral anticoagulant or aspirin for nonvalvular atrial fibrillation with CHA2DS2-VASc (Congestive heart failure, Hypertension, Age [> 65 y, 1 point; > 75 y, 2 points], Diabetes, previous Stroke/transient ischemic attack [2 points]) score of 1.21 The ACC/ AHA guidelines were updated in 2023 to recommend against antiplatelet therapy as an alternative to anticoagulation for reducing cardioembolic stroke risk among patients with atrial fibrillation with no indication for antiplatelet therapy because of risk of harm.22 If a patient has no risk factors for stroke, aspirin is not recommended to prevent thromboembolic events because of a lack of benefit. Interventions from this quality improvement study were completed before the 2023 atrial fibrillation guideline was published and therefore in this study aspirin was not discontinued when used for atrial fibrillation. Aspirin use for atrial fibrillation might benefit from similar discontinuation efforts analyzed within this study. Beyond atrial fibrillation, major guidelines do not comment on the use of aspirin for any other indications in the absence of clinical ASCVD.
Limitations
This study is limited by the lack of clinical consensus for complex patients and demonstrates the importance of individualized patient assessment when considering discontinuing aspirin. Because of the project’s relatively short intervention period, aspirin deprescribing rates could decrease over time and repeated education efforts might be necessary to see lasting impact. Health care professionals from services outside of primary care also might have discontinued aspirin during the study period unrelated to the education and these discontinued aspirin prescriptions could contribute to the higher rate observed among PCPs. This study included a specific population cohort of male, US veterans and might not reflect other populations where these interventions could be implemented.
The measurement of time spent by pharmacists and PCPs is an additional limitation. Although it is expected that PCPs attempt to discontinue aspirin during their existing patient care appointments, the time spent during visits was not measured or documented. Direct deprescribing by pharmacist CPRS review required a significant amount of time and could be a barrier to successful intervention by CPPs in patient aligned care teams.
To reduce the time pharmacists spent completing CPRS reviews, an aspirin deprescribing clinical reminder tool could be used to assess use and appropriate indication quickly during any primary care visit led by a PCP or CPP. In addition, it is recommended that pharmacists regularly educate health care professionals on guideline recommendations for aspirin use among geriatric patients. Future studies of the incidence of major cardiovascular events after aspirin deprescribing among geriatric patients and a longitudinal cost/benefit analysis could support these initiatives.
Conclusions
In this study, pharmacists successfully deprescribed inappropriate medications, such as aspirin. However, pharmacist-led PCP education is more efficient compared with direct deprescribing using a population-level review. PCP education requires less time and could allow ambulatory care pharmacists to spend more time on other direct patient care interventions to improve quality and access to care in primary care clinics. This study’s results further support the role of pharmacists in deprescribing PIMs for older adults and the use of a deprescribing tool, such as VIONE, in a primary care setting.
- US Preventive Services Task Force; Davidson KW, Barry MJ, et al. Aspirin use to prevent cardiovascular disease: US Preventive Services Task Force recommendation statement. JAMA. 2022;327(16):1577-1584. doi:10.1001/jama.2022.4983
- McNeil JJ, Nelson MR, Woods RL, et al. Effect of aspirin on all-cause mortality in the healthy elderly. N Engl J Med. 2018;379(16):1519-1528. doi:10.1056/NEJMoa1803955
- Barry MJ, Wolff TA, Pbert L, et al. Putting evidence into practice: an update on the US Preventive Services Task Force methods for developing recommendations for preventive services. Ann Fam Med. 2023;21(2):165-171. doi:10.1370/afm.2946
- Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/ AHA Guideline on the Primary Prevention of Cardiovascular Disease: A report of the American College of Cardiology/ American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;140(11):e596-e646. doi:10.1161/CIR.0000000000000678
- American Diabetes Association Professional Practice Committee. 10. Cardiovascular disease and risk management: Standards of care in diabetes-2024. Diabetes Care. 2024;47(Suppl 1):S179-S218. doi:10.2337/dc24-S010
- Ong SY, Chui P, Bhargava A, Justice A, Hauser RG. Estimating aspirin overuse for primary prevention of atherosclerotic cardiovascular disease (from a nationwide healthcare system). Am J Cardiol. 2020;137:25-30. doi:10.1016/j.amjcard.2020.09.042
- Weiss AJ, Jiang HJ. Overview of clinical conditions with frequent and costly hospital readmissions by payer, 2018. In: Healthcare Cost and Utilization Project (HCUP) Statistical Briefs. Agency for Healthcare Research and Quality (US); July 20, 2021.
- Krishnaswami A, Steinman MA, Goyal P, et al. Deprescribing in older adults with cardiovascular disease. J Am Coll Cardiol. 2019;73(20):2584-2595. doi:10.1016/j.jacc.2019.03.467
- Association of American Medical Colleges. The complexities of physician supply and demand: projections from 2019 to 2034. Accessed March 17, 2024. https://www.aamc.org/media/54681/download
- US Department of Veterans Affairs, Veterans Health Administration. VHA Directive 1108.07(1): General pharmacy service requirements. November 28, 2022. Accessed March 17, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=10045
- US Department of Veterans Affairs, Veterans Health Administration. VHA Handbook 1108.11(3): Clinical pharmacy services. July 1, 2015. Accessed March 17, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=3120
- US Department of Veterans Affairs. Clinical pharmacist practitioner (CPP) to improve access to and quality of care August 2021. August 2021. Accessed May 19, 2023. https://www.pbm.va.gov/PBM/CPPO/Documents/ExternalFactSheet_OptimizingtheCPPToImproveAccess_508.pdf
- Ammerman CA, Simpkins BA, Warman N, Downs TN. Potentially inappropriate medications in older adults: Deprescribing with a clinical pharmacist. J Am Geriatr Soc. 2019;67(1):115-118. doi:10.1111/jgs.15623
- Rothbauer K, Siodlak M, Dreischmeier E, Ranola TS, Welch L. Evaluation of a pharmacist-driven ambulatory aspirin deprescribing protocol. Fed Pract. 2022;39(suppl 5):S37- S41a. doi:10.12788/fp.0294
- US Department of Veterans Affairs. VIONE changes the way VA handles prescriptions. January 25, 2020. Accessed May 21, 2023. https://news.va.gov/70709/vione-changes-way-va-handles-prescriptions/
- 2023 American Geriatrics Society Beers Criteria Update Expert Panel. American Geriatrics Society 2023 updated AGS Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2023;71(7):2052- 2081. doi:10.1111/jgs.18372
- O’Mahony D, Cherubini A, Guiteras AR, et al. STOPP/ START criteria for potentially inappropriate prescribing in older people: version 3. Eur Geriatr Med. 2023;14(4):625- 632. doi:10.1007/s41999-023-00777-y
- Draeger C, Lodhi F, Geissinger N, Larson T, Griesbach S. Interdisciplinary deprescribing of aspirin through prescriber education and provision of patient-specific recommendations. WMJ. 2022;121(3):220-225
- de Lusignan S, Hinton W, Seidu S, et al. Dashboards to reduce inappropriate prescribing of metformin and aspirin: A quality assurance programme in a primary care sentinel network. Prim Care Diabetes. 2021;15(6):1075-1079. doi:10.1016/j.pcd.2021.06.003
- Nelson MW, Downs TN, Puglisi GM, Simpkins BA, Collier AS. Use of a deprescribing tool in an interdisciplinary primary-care patient-aligned care team. Sr Care Pharm. 2022;37(1):34-43. doi:10.4140/TCP.n.2022.34
- January CT, Wann LS, Alpert JS, et al. 2014 AHA/ ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the Heart Rhythm Society. Circulation. 2014;130(23):e199-e267. doi:10.1161/CIR.0000000000000041
- Joglar JA, Chung MK, Armbruster AL, et al. 2023 ACC/ AHA/ACCP/HRS Guideline for the Diagnosis and Management of Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines Circulation. 2024;149(1):e1- e156. doi:10.1161/CIR.0000000000001193
- US Preventive Services Task Force; Davidson KW, Barry MJ, et al. Aspirin use to prevent cardiovascular disease: US Preventive Services Task Force recommendation statement. JAMA. 2022;327(16):1577-1584. doi:10.1001/jama.2022.4983
- McNeil JJ, Nelson MR, Woods RL, et al. Effect of aspirin on all-cause mortality in the healthy elderly. N Engl J Med. 2018;379(16):1519-1528. doi:10.1056/NEJMoa1803955
- Barry MJ, Wolff TA, Pbert L, et al. Putting evidence into practice: an update on the US Preventive Services Task Force methods for developing recommendations for preventive services. Ann Fam Med. 2023;21(2):165-171. doi:10.1370/afm.2946
- Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/ AHA Guideline on the Primary Prevention of Cardiovascular Disease: A report of the American College of Cardiology/ American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;140(11):e596-e646. doi:10.1161/CIR.0000000000000678
- American Diabetes Association Professional Practice Committee. 10. Cardiovascular disease and risk management: Standards of care in diabetes-2024. Diabetes Care. 2024;47(Suppl 1):S179-S218. doi:10.2337/dc24-S010
- Ong SY, Chui P, Bhargava A, Justice A, Hauser RG. Estimating aspirin overuse for primary prevention of atherosclerotic cardiovascular disease (from a nationwide healthcare system). Am J Cardiol. 2020;137:25-30. doi:10.1016/j.amjcard.2020.09.042
- Weiss AJ, Jiang HJ. Overview of clinical conditions with frequent and costly hospital readmissions by payer, 2018. In: Healthcare Cost and Utilization Project (HCUP) Statistical Briefs. Agency for Healthcare Research and Quality (US); July 20, 2021.
- Krishnaswami A, Steinman MA, Goyal P, et al. Deprescribing in older adults with cardiovascular disease. J Am Coll Cardiol. 2019;73(20):2584-2595. doi:10.1016/j.jacc.2019.03.467
- Association of American Medical Colleges. The complexities of physician supply and demand: projections from 2019 to 2034. Accessed March 17, 2024. https://www.aamc.org/media/54681/download
- US Department of Veterans Affairs, Veterans Health Administration. VHA Directive 1108.07(1): General pharmacy service requirements. November 28, 2022. Accessed March 17, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=10045
- US Department of Veterans Affairs, Veterans Health Administration. VHA Handbook 1108.11(3): Clinical pharmacy services. July 1, 2015. Accessed March 17, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=3120
- US Department of Veterans Affairs. Clinical pharmacist practitioner (CPP) to improve access to and quality of care August 2021. August 2021. Accessed May 19, 2023. https://www.pbm.va.gov/PBM/CPPO/Documents/ExternalFactSheet_OptimizingtheCPPToImproveAccess_508.pdf
- Ammerman CA, Simpkins BA, Warman N, Downs TN. Potentially inappropriate medications in older adults: Deprescribing with a clinical pharmacist. J Am Geriatr Soc. 2019;67(1):115-118. doi:10.1111/jgs.15623
- Rothbauer K, Siodlak M, Dreischmeier E, Ranola TS, Welch L. Evaluation of a pharmacist-driven ambulatory aspirin deprescribing protocol. Fed Pract. 2022;39(suppl 5):S37- S41a. doi:10.12788/fp.0294
- US Department of Veterans Affairs. VIONE changes the way VA handles prescriptions. January 25, 2020. Accessed May 21, 2023. https://news.va.gov/70709/vione-changes-way-va-handles-prescriptions/
- 2023 American Geriatrics Society Beers Criteria Update Expert Panel. American Geriatrics Society 2023 updated AGS Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2023;71(7):2052- 2081. doi:10.1111/jgs.18372
- O’Mahony D, Cherubini A, Guiteras AR, et al. STOPP/ START criteria for potentially inappropriate prescribing in older people: version 3. Eur Geriatr Med. 2023;14(4):625- 632. doi:10.1007/s41999-023-00777-y
- Draeger C, Lodhi F, Geissinger N, Larson T, Griesbach S. Interdisciplinary deprescribing of aspirin through prescriber education and provision of patient-specific recommendations. WMJ. 2022;121(3):220-225
- de Lusignan S, Hinton W, Seidu S, et al. Dashboards to reduce inappropriate prescribing of metformin and aspirin: A quality assurance programme in a primary care sentinel network. Prim Care Diabetes. 2021;15(6):1075-1079. doi:10.1016/j.pcd.2021.06.003
- Nelson MW, Downs TN, Puglisi GM, Simpkins BA, Collier AS. Use of a deprescribing tool in an interdisciplinary primary-care patient-aligned care team. Sr Care Pharm. 2022;37(1):34-43. doi:10.4140/TCP.n.2022.34
- January CT, Wann LS, Alpert JS, et al. 2014 AHA/ ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the Heart Rhythm Society. Circulation. 2014;130(23):e199-e267. doi:10.1161/CIR.0000000000000041
- Joglar JA, Chung MK, Armbruster AL, et al. 2023 ACC/ AHA/ACCP/HRS Guideline for the Diagnosis and Management of Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines Circulation. 2024;149(1):e1- e156. doi:10.1161/CIR.0000000000001193
Pharmacist-Led Deprescribing of Aspirin for Primary Prevention of Cardiovascular Disease Among Geriatric Veterans
Pharmacist-Led Deprescribing of Aspirin for Primary Prevention of Cardiovascular Disease Among Geriatric Veterans