Theme
medstat_icymi_bcell
icymibcell
Main menu
ICYMI B-Cell Lymphoma Featured Menu
Unpublish
Altmetric
Click for Credit Button Label
Click For Credit
DSM Affiliated
Display in offset block
Enable Disqus
Display Author and Disclosure Link
Publication Type
News
Slot System
Featured Buckets
Disable Sticky Ads
Disable Ad Block Mitigation
Featured Buckets Admin
Show Ads on this Publication's Homepage
Consolidated Pub
Show Article Page Numbers on TOC
Use larger logo size
Off
publication_blueconic_enabled
Off
Show More Destinations Menu
Disable Adhesion on Publication
Off
Restore Menu Label on Mobile Navigation
Disable Facebook Pixel from Publication
Exclude this publication from publication selection on articles and quiz
Challenge Center
Disable Inline Native ads
Activity Salesforce Deliverable ID
343187.19
Activity ID
95012
Product Name
Clinical Briefings ICYMI
Product ID
112

Pembrolizumab Dose Conversion Adoption and Immune-Mediated Adverse Events

Article Type
Changed
Thu, 12/15/2022 - 14:36

Background/Purpose 

On April 28, 2020, the Food and Drug Administration approved pembrolizumab 400mg intravenous (IV) every 6 weeks. This dosing update was rapidly adopted by VA Northeast Ohio Healthcare System (VANEOHS) hematology/oncology providers to minimize infusion appointments, for patient convenience and COVID precautions. On May 1, 2020, pembrolizumab order set templates were updated to reflect the extended interval dosing, however providers are still able to change orders to 200mg IV every 3 weeks if needed. Due to administration of higher pembrolizumab doses, there could be increased development of immune-mediated adverse events (IrAEs). This review quantified the clinic visits saved at VANEOHS by adoption of pembrolizumab 400mg dosing and report adverse events that resulted in pembrolizumab dose reduction.

Methods 

A report of all pembrolizumab orders from May 1, 2020 to May 1, 2021 was obtained. All pembrolizumab 200mg orders were reviewed to evaluate reasoning for the use of the 200mg dose. A retrospective chart review was performed for patients who required a pembrolizumab dose reduction to evaluate safety. Descriptive statistics were used.

Results 

There was a total of 277 pembrolizumab orders from May 1, 2020 to May 1, 2021. Of these orders, 211 (76%) were converted to pembrolizumab 400mg IV every 6 weeks, while 66 (24%) orders remained at pembrolizumab 200mg IV every 3 weeks. It is estimated that there were 211 infusion appointments avoided due to the conversion to pembrolizumab 400mg IV every 6-week dosing. The 277 pembrolizumab orders were used to treat 77 unique patients. Eighteen patients continued to receive pembrolizumab 200mg following the conversion. Sixteen of these patients were maintained on pembrolizumab 200mg due to concomitant chemotherapy schedules. One patient was receiving pembrolizumab 200mg based on clinical trial dosing. One patient returned to pembrolizumab 200mg due to an increase in drainage from pleurx catheter while receiving 400mg dose.

Implications

The conversion from pembrolizumab 200mg every 3 weeks to pembrolizumab 400mg every 6 weeks avoided approximately 200 infusion appointments without an increase in safety concerns. This supporting data may aid in supporting extended interval dosing of other immunotherapy agents.

Author and Disclosure Information

Louis Stokes Cleveland VA Medical Center

Issue
Federal Practitioner - 38(4)s
Publications
Topics
Page Number
S12
Sections
Author and Disclosure Information

Louis Stokes Cleveland VA Medical Center

Author and Disclosure Information

Louis Stokes Cleveland VA Medical Center

Background/Purpose 

On April 28, 2020, the Food and Drug Administration approved pembrolizumab 400mg intravenous (IV) every 6 weeks. This dosing update was rapidly adopted by VA Northeast Ohio Healthcare System (VANEOHS) hematology/oncology providers to minimize infusion appointments, for patient convenience and COVID precautions. On May 1, 2020, pembrolizumab order set templates were updated to reflect the extended interval dosing, however providers are still able to change orders to 200mg IV every 3 weeks if needed. Due to administration of higher pembrolizumab doses, there could be increased development of immune-mediated adverse events (IrAEs). This review quantified the clinic visits saved at VANEOHS by adoption of pembrolizumab 400mg dosing and report adverse events that resulted in pembrolizumab dose reduction.

Methods 

A report of all pembrolizumab orders from May 1, 2020 to May 1, 2021 was obtained. All pembrolizumab 200mg orders were reviewed to evaluate reasoning for the use of the 200mg dose. A retrospective chart review was performed for patients who required a pembrolizumab dose reduction to evaluate safety. Descriptive statistics were used.

Results 

There was a total of 277 pembrolizumab orders from May 1, 2020 to May 1, 2021. Of these orders, 211 (76%) were converted to pembrolizumab 400mg IV every 6 weeks, while 66 (24%) orders remained at pembrolizumab 200mg IV every 3 weeks. It is estimated that there were 211 infusion appointments avoided due to the conversion to pembrolizumab 400mg IV every 6-week dosing. The 277 pembrolizumab orders were used to treat 77 unique patients. Eighteen patients continued to receive pembrolizumab 200mg following the conversion. Sixteen of these patients were maintained on pembrolizumab 200mg due to concomitant chemotherapy schedules. One patient was receiving pembrolizumab 200mg based on clinical trial dosing. One patient returned to pembrolizumab 200mg due to an increase in drainage from pleurx catheter while receiving 400mg dose.

Implications

The conversion from pembrolizumab 200mg every 3 weeks to pembrolizumab 400mg every 6 weeks avoided approximately 200 infusion appointments without an increase in safety concerns. This supporting data may aid in supporting extended interval dosing of other immunotherapy agents.

Background/Purpose 

On April 28, 2020, the Food and Drug Administration approved pembrolizumab 400mg intravenous (IV) every 6 weeks. This dosing update was rapidly adopted by VA Northeast Ohio Healthcare System (VANEOHS) hematology/oncology providers to minimize infusion appointments, for patient convenience and COVID precautions. On May 1, 2020, pembrolizumab order set templates were updated to reflect the extended interval dosing, however providers are still able to change orders to 200mg IV every 3 weeks if needed. Due to administration of higher pembrolizumab doses, there could be increased development of immune-mediated adverse events (IrAEs). This review quantified the clinic visits saved at VANEOHS by adoption of pembrolizumab 400mg dosing and report adverse events that resulted in pembrolizumab dose reduction.

Methods 

A report of all pembrolizumab orders from May 1, 2020 to May 1, 2021 was obtained. All pembrolizumab 200mg orders were reviewed to evaluate reasoning for the use of the 200mg dose. A retrospective chart review was performed for patients who required a pembrolizumab dose reduction to evaluate safety. Descriptive statistics were used.

Results 

There was a total of 277 pembrolizumab orders from May 1, 2020 to May 1, 2021. Of these orders, 211 (76%) were converted to pembrolizumab 400mg IV every 6 weeks, while 66 (24%) orders remained at pembrolizumab 200mg IV every 3 weeks. It is estimated that there were 211 infusion appointments avoided due to the conversion to pembrolizumab 400mg IV every 6-week dosing. The 277 pembrolizumab orders were used to treat 77 unique patients. Eighteen patients continued to receive pembrolizumab 200mg following the conversion. Sixteen of these patients were maintained on pembrolizumab 200mg due to concomitant chemotherapy schedules. One patient was receiving pembrolizumab 200mg based on clinical trial dosing. One patient returned to pembrolizumab 200mg due to an increase in drainage from pleurx catheter while receiving 400mg dose.

Implications

The conversion from pembrolizumab 200mg every 3 weeks to pembrolizumab 400mg every 6 weeks avoided approximately 200 infusion appointments without an increase in safety concerns. This supporting data may aid in supporting extended interval dosing of other immunotherapy agents.

Issue
Federal Practitioner - 38(4)s
Issue
Federal Practitioner - 38(4)s
Page Number
S12
Page Number
S12
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 10/01/2021 - 14:30
Un-Gate On Date
Fri, 10/01/2021 - 14:30
Use ProPublica
CFC Schedule Remove Status
Fri, 10/01/2021 - 14:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Novel and Alternative Strategies for Management of Panitumumab-Induced Hypomagnesemia

Article Type
Changed
Thu, 12/15/2022 - 14:36

Background

Panitumumab is an epidermal growth factor receptor (EGFR) inhibiting monoclonal antibody approved for the treatment of RAS wild-type metastatic colorectal cancer (mCRC), which has an incidence of hypomagnesemia of approximately 35%. Grade 3 or 4 hypomagnesemia occurs in roughly 7% of patients, which can lead to serious complications such as seizures and arrhythmias. In one study, hypomagnesemia led to discontinuation of targeted therapy in 3% of patients. Currently, there is no standardized prophylactic strategy or treatment protocol for panitumumab-induced hypomagnesemia. In cases of refractory hypomagnesemia, it is recommended to discontinue panitumumab, even if the patient is deriving clinical benefit.

 

Case Report

This 59-year-old male was diagnosed with RAS wild-type mCRC and had already progressed through multiple lines of treatment. Panitumumab was initiated with good response; however, the drug was discontinued due to grade 4 hypomagnesemia, despite intravenous and oral supplementation. As the patient progressed through further lines of treatment, the decision was made to retry panitumumab. Grade 2-3 hypomagnesemia persisted throughout treatment, requiring frequent magnesium infusions. Innovative and alternative treatment options were investigated in an effort to improve his quality of life. In addition to oral and intravenous magnesium replacement, an ambulatory elastomeric pump, traditionally used for fluorouracil administration, was repurposed to deliver between 6 and 24 grams of magnesium sulfate over 24 to 72 hours. The pump was generally well tolerated with the exception of mild skin irritation around the port site, which prevented a transition to longer infusion times. The ambulatory elastomeric pump decreased the frequency of healthcare visits and improved the hypomagnesemia sufficiently to continue treatment with panitumumab, although levels did not fully normalize. A two-week trial of amiloride was also attempted to decrease renal magnesium wasting. Amiloride normalized magnesium levels but had to be discontinued due to asymptomatic hyperkalemia. This case report suggests that amiloride and magnesium replacement via ambulatory elastomeric pumps may be safe and effective treatment options for panitumumab-induced refractory hypomagnesemia in mCRC, potentially improving quality of life and allowing beneficial anti-cancer treatments to continue. Future studies should further evaluate optimization of amiloride and intravenous magnesium replacement via ambulatory elastomeric pump.

Author and Disclosure Information

South Texas Veterans Health Administration, University of Texas at Austin College of Pharmacy

Issue
Federal Practitioner - 38(4)s
Publications
Topics
Page Number
S12
Sections
Author and Disclosure Information

South Texas Veterans Health Administration, University of Texas at Austin College of Pharmacy

Author and Disclosure Information

South Texas Veterans Health Administration, University of Texas at Austin College of Pharmacy

Background

Panitumumab is an epidermal growth factor receptor (EGFR) inhibiting monoclonal antibody approved for the treatment of RAS wild-type metastatic colorectal cancer (mCRC), which has an incidence of hypomagnesemia of approximately 35%. Grade 3 or 4 hypomagnesemia occurs in roughly 7% of patients, which can lead to serious complications such as seizures and arrhythmias. In one study, hypomagnesemia led to discontinuation of targeted therapy in 3% of patients. Currently, there is no standardized prophylactic strategy or treatment protocol for panitumumab-induced hypomagnesemia. In cases of refractory hypomagnesemia, it is recommended to discontinue panitumumab, even if the patient is deriving clinical benefit.

 

Case Report

This 59-year-old male was diagnosed with RAS wild-type mCRC and had already progressed through multiple lines of treatment. Panitumumab was initiated with good response; however, the drug was discontinued due to grade 4 hypomagnesemia, despite intravenous and oral supplementation. As the patient progressed through further lines of treatment, the decision was made to retry panitumumab. Grade 2-3 hypomagnesemia persisted throughout treatment, requiring frequent magnesium infusions. Innovative and alternative treatment options were investigated in an effort to improve his quality of life. In addition to oral and intravenous magnesium replacement, an ambulatory elastomeric pump, traditionally used for fluorouracil administration, was repurposed to deliver between 6 and 24 grams of magnesium sulfate over 24 to 72 hours. The pump was generally well tolerated with the exception of mild skin irritation around the port site, which prevented a transition to longer infusion times. The ambulatory elastomeric pump decreased the frequency of healthcare visits and improved the hypomagnesemia sufficiently to continue treatment with panitumumab, although levels did not fully normalize. A two-week trial of amiloride was also attempted to decrease renal magnesium wasting. Amiloride normalized magnesium levels but had to be discontinued due to asymptomatic hyperkalemia. This case report suggests that amiloride and magnesium replacement via ambulatory elastomeric pumps may be safe and effective treatment options for panitumumab-induced refractory hypomagnesemia in mCRC, potentially improving quality of life and allowing beneficial anti-cancer treatments to continue. Future studies should further evaluate optimization of amiloride and intravenous magnesium replacement via ambulatory elastomeric pump.

Background

Panitumumab is an epidermal growth factor receptor (EGFR) inhibiting monoclonal antibody approved for the treatment of RAS wild-type metastatic colorectal cancer (mCRC), which has an incidence of hypomagnesemia of approximately 35%. Grade 3 or 4 hypomagnesemia occurs in roughly 7% of patients, which can lead to serious complications such as seizures and arrhythmias. In one study, hypomagnesemia led to discontinuation of targeted therapy in 3% of patients. Currently, there is no standardized prophylactic strategy or treatment protocol for panitumumab-induced hypomagnesemia. In cases of refractory hypomagnesemia, it is recommended to discontinue panitumumab, even if the patient is deriving clinical benefit.

 

Case Report

This 59-year-old male was diagnosed with RAS wild-type mCRC and had already progressed through multiple lines of treatment. Panitumumab was initiated with good response; however, the drug was discontinued due to grade 4 hypomagnesemia, despite intravenous and oral supplementation. As the patient progressed through further lines of treatment, the decision was made to retry panitumumab. Grade 2-3 hypomagnesemia persisted throughout treatment, requiring frequent magnesium infusions. Innovative and alternative treatment options were investigated in an effort to improve his quality of life. In addition to oral and intravenous magnesium replacement, an ambulatory elastomeric pump, traditionally used for fluorouracil administration, was repurposed to deliver between 6 and 24 grams of magnesium sulfate over 24 to 72 hours. The pump was generally well tolerated with the exception of mild skin irritation around the port site, which prevented a transition to longer infusion times. The ambulatory elastomeric pump decreased the frequency of healthcare visits and improved the hypomagnesemia sufficiently to continue treatment with panitumumab, although levels did not fully normalize. A two-week trial of amiloride was also attempted to decrease renal magnesium wasting. Amiloride normalized magnesium levels but had to be discontinued due to asymptomatic hyperkalemia. This case report suggests that amiloride and magnesium replacement via ambulatory elastomeric pumps may be safe and effective treatment options for panitumumab-induced refractory hypomagnesemia in mCRC, potentially improving quality of life and allowing beneficial anti-cancer treatments to continue. Future studies should further evaluate optimization of amiloride and intravenous magnesium replacement via ambulatory elastomeric pump.

Issue
Federal Practitioner - 38(4)s
Issue
Federal Practitioner - 38(4)s
Page Number
S12
Page Number
S12
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 10/01/2021 - 14:30
Un-Gate On Date
Fri, 10/01/2021 - 14:30
Use ProPublica
CFC Schedule Remove Status
Fri, 10/01/2021 - 14:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

A Single-Center Experience of Cardiac-related Adverse Events from Immune Checkpoint Inhibitors

Article Type
Changed
Thu, 12/15/2022 - 14:36

Introduction

There have been incident reports of cardiac-related adverse events (CrAE) from immune checkpoint inhibitors (ICPI); however, the true incidence and subsequent management of these potential side effects have not been defined. It is therefore important to study ICPI related cardiac dysfunction to assist in monitoring and surveillance of these patients.

Methods

63 patients who received nivolumab and pembrolizumab at Stratton VAMC Albany between January 2015 to December 2018 were studied. Retrospective chart review was done to identify the CrAE up to two-year post-therapy completion or discontinuation. Naranjo score was used to assess drug-related side effect. IRB approval was obtained.

Results 

CrAE were defined as new onset arrythmia identified on electrocardiogram, evidence of cardiomyopathy on echocardiogram, an acute coronary event, and hospitalizations from primary cardiac disorder following ICPI administration. Of the 63 patients, 6 patients developed CrAE. Our review showed 3 patients developed new arrythmias including 1 with atrial fibrillation, and 2 with atrial flutter. There was 1 case each of new heart failure with reduced ejection fraction and pericarditis with pericardial tamponade. 1 patient developed acute coronary syndrome in addition to complete heart block. Of the 6 patients, 2 had elevated brain natriuretic peptide (BNP) prior to onset of CrAE. Elevated markers including BNP and troponin-I were also seen in 13 patients with preexisting heart conditions without CrAE. Duration of therapy was variable for all patients with CrAE. Therapy was continued for 3 patients without recurrence of CrAE. Therapy was permanently discontinued in the patient who developed pericardial effusion (grade IV toxicity). The remaining 2 patients had additional concurrent immune-related toxicities that required discontinuation of therapy. Our analysis showed 25/63 patients with pre-existing cardiac conditions (including arrhythmia, heart failure or coronary artery disease) who did not develop new CrAE; however 6 of these patients required hospitalization for exacerbation related to these pre-existing conditions.

 

Conclusions

CrAE can occur with ICPIs, and vigilance is required in high-risk patient including those with pre-existing cardiac comorbidity. Further studies are required to establish if baseline screening EKG and echocardiogram should be obtained for all patients starting ICPI.

Author and Disclosure Information

Albany Medical Center, Albany Stratton VA Medical Center

Issue
Federal Practitioner - 38(4)s
Publications
Topics
Page Number
S11
Sections
Author and Disclosure Information

Albany Medical Center, Albany Stratton VA Medical Center

Author and Disclosure Information

Albany Medical Center, Albany Stratton VA Medical Center

Introduction

There have been incident reports of cardiac-related adverse events (CrAE) from immune checkpoint inhibitors (ICPI); however, the true incidence and subsequent management of these potential side effects have not been defined. It is therefore important to study ICPI related cardiac dysfunction to assist in monitoring and surveillance of these patients.

Methods

63 patients who received nivolumab and pembrolizumab at Stratton VAMC Albany between January 2015 to December 2018 were studied. Retrospective chart review was done to identify the CrAE up to two-year post-therapy completion or discontinuation. Naranjo score was used to assess drug-related side effect. IRB approval was obtained.

Results 

CrAE were defined as new onset arrythmia identified on electrocardiogram, evidence of cardiomyopathy on echocardiogram, an acute coronary event, and hospitalizations from primary cardiac disorder following ICPI administration. Of the 63 patients, 6 patients developed CrAE. Our review showed 3 patients developed new arrythmias including 1 with atrial fibrillation, and 2 with atrial flutter. There was 1 case each of new heart failure with reduced ejection fraction and pericarditis with pericardial tamponade. 1 patient developed acute coronary syndrome in addition to complete heart block. Of the 6 patients, 2 had elevated brain natriuretic peptide (BNP) prior to onset of CrAE. Elevated markers including BNP and troponin-I were also seen in 13 patients with preexisting heart conditions without CrAE. Duration of therapy was variable for all patients with CrAE. Therapy was continued for 3 patients without recurrence of CrAE. Therapy was permanently discontinued in the patient who developed pericardial effusion (grade IV toxicity). The remaining 2 patients had additional concurrent immune-related toxicities that required discontinuation of therapy. Our analysis showed 25/63 patients with pre-existing cardiac conditions (including arrhythmia, heart failure or coronary artery disease) who did not develop new CrAE; however 6 of these patients required hospitalization for exacerbation related to these pre-existing conditions.

 

Conclusions

CrAE can occur with ICPIs, and vigilance is required in high-risk patient including those with pre-existing cardiac comorbidity. Further studies are required to establish if baseline screening EKG and echocardiogram should be obtained for all patients starting ICPI.

Introduction

There have been incident reports of cardiac-related adverse events (CrAE) from immune checkpoint inhibitors (ICPI); however, the true incidence and subsequent management of these potential side effects have not been defined. It is therefore important to study ICPI related cardiac dysfunction to assist in monitoring and surveillance of these patients.

Methods

63 patients who received nivolumab and pembrolizumab at Stratton VAMC Albany between January 2015 to December 2018 were studied. Retrospective chart review was done to identify the CrAE up to two-year post-therapy completion or discontinuation. Naranjo score was used to assess drug-related side effect. IRB approval was obtained.

Results 

CrAE were defined as new onset arrythmia identified on electrocardiogram, evidence of cardiomyopathy on echocardiogram, an acute coronary event, and hospitalizations from primary cardiac disorder following ICPI administration. Of the 63 patients, 6 patients developed CrAE. Our review showed 3 patients developed new arrythmias including 1 with atrial fibrillation, and 2 with atrial flutter. There was 1 case each of new heart failure with reduced ejection fraction and pericarditis with pericardial tamponade. 1 patient developed acute coronary syndrome in addition to complete heart block. Of the 6 patients, 2 had elevated brain natriuretic peptide (BNP) prior to onset of CrAE. Elevated markers including BNP and troponin-I were also seen in 13 patients with preexisting heart conditions without CrAE. Duration of therapy was variable for all patients with CrAE. Therapy was continued for 3 patients without recurrence of CrAE. Therapy was permanently discontinued in the patient who developed pericardial effusion (grade IV toxicity). The remaining 2 patients had additional concurrent immune-related toxicities that required discontinuation of therapy. Our analysis showed 25/63 patients with pre-existing cardiac conditions (including arrhythmia, heart failure or coronary artery disease) who did not develop new CrAE; however 6 of these patients required hospitalization for exacerbation related to these pre-existing conditions.

 

Conclusions

CrAE can occur with ICPIs, and vigilance is required in high-risk patient including those with pre-existing cardiac comorbidity. Further studies are required to establish if baseline screening EKG and echocardiogram should be obtained for all patients starting ICPI.

Issue
Federal Practitioner - 38(4)s
Issue
Federal Practitioner - 38(4)s
Page Number
S11
Page Number
S11
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 10/01/2021 - 14:15
Un-Gate On Date
Fri, 10/01/2021 - 14:15
Use ProPublica
CFC Schedule Remove Status
Fri, 10/01/2021 - 14:15
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

VA Turns to Telehealth to Address Delays in Genetic Counseling

Article Type
Changed
Thu, 12/15/2022 - 14:36
Challenges remain as VA is understaffed for the demand and commercial laboratories are not equipped to handle complex veteran cases.

The U.S. Department of Veteran Affairs (VA) has been unable to provide genetic counseling to veterans at the same level as the civilian community, and other gaps exist, a genetic counselor told oncologist and hematologist colleagues. The good news is that telemedicine is turning out to be a valuable and proven way to reach veterans who need this kind of care, she said, although certain patients are being left behind.

 

“To me, telehealth is no doubt the way to go. But it is really important that we continue to look into these disparities, what's causing them, and how we can find a path forward,” said

Deborah Hartzfeld, MS, CGC, of the Genomic Medicine Service based in Salt Lake City, Utah. She spoke in a presentation at the 2021 annual meeting of the Association of VA Hematology/Oncology (AVAHO) that was held virtually and in person in Denver, Colorado, from September 24 to September 26, 2021.

 

As Harzfeld explained, the genetic counselor workforce is expanding along with the number of indications for genetic testing, especially in cancer, “where the need for germline genetic testing for inherited cancer genes becomes broader every year.”

 

Genetic counselors are a homogenous group, she said, as revealed by a 2021 survey of most of the nation’s 5,629 certified generic counselors. The North American survey, by the National Society of Genetic Counselors, found that 94% of respondents identified themselves as female, and 90% were white/non-Hispanic.  

 

The survey report also noted that “the genetic counseling profession has grown by over 100% in the last 10 years and is expected to grow another 100% over the next 10 years. By 2025 there should be nearly 7,500 certified genetic counselors, and by 2030 there are likely to be over 10,000.”

 

Genetic counseling within the VA has also grown rapidly. In 2010, Harzfeld said, about 737 veterans were referred for the service. In 2020, the number was about 10,000, with about half referred for personal or family history of cancer.

 

The VA has 18 genetic counselors, not all of whom are actively seeing patients or working full time, she said. “Per the National Society of Genetic Counselors, there's one clinical genetic counselor per 100,000 people in the general population,” she said. “It's one for about 474,000 in the VA.”

 

Wait times for genetic counseling within the VA exceed Mission Act standards outside of urgent referrals in matters such as surgical or medical management, she said. “We usually see those patients within a week, but other folks have to wait or are referred into the community. It remains unclear how many of our patients could access care easily in the community or what the wait times at any individual VA will be.”

 

Fortunately, she said, telemedicine has increased access to genetic counseling within the general population and the VA, Harzfeld said. “A recent systematic evidence review found providing genetic counseling via video or telephone is comparable to in-person care, it increases access and it's likely feasible and acceptable to major stakeholders. It's worth noting that the data in this evidence review was collected prior to COVID-19 when fewer programs were using telehealth.”

 

Genetic counseling works especially well via telehealth because counselors don’t perform physical examinations, she said. “Prior to COVID, service probably saw maybe 4 VVC [VA Video Connect] appointments per month for genetic counseling. Now, VVC makes up about 70% of our new patient encounters. About 25% are telephone and about 5% are clinical video telehealth where the veteran goes into their clinic to be seated in front of the machine.”

 

Research has suggested that non-White patients are 40 to 50% less likely to be referred to telehealth for genetic counseling vs. in-person encounters, she said, although women in general (including black women) are more likely to be referred.

 

Harzfeld highlighted several challenges facing genetic counseling in the VA. She notes that contracted laboratories aren’t “really set up to be experts in germline genetic testing, so they’re not as nimble, and their test catalogs are not most likely going to be as comprehensive enough for what is needed.” Also, she said, “test ordering can be quite burdensome.”

 

“We need to continue working with various partners to increase access and the ease of ordering genetic testing,” she said.

 

Hartzfeld reports no disclosures.

 

Publications
Topics
Sections
Challenges remain as VA is understaffed for the demand and commercial laboratories are not equipped to handle complex veteran cases.
Challenges remain as VA is understaffed for the demand and commercial laboratories are not equipped to handle complex veteran cases.

The U.S. Department of Veteran Affairs (VA) has been unable to provide genetic counseling to veterans at the same level as the civilian community, and other gaps exist, a genetic counselor told oncologist and hematologist colleagues. The good news is that telemedicine is turning out to be a valuable and proven way to reach veterans who need this kind of care, she said, although certain patients are being left behind.

 

“To me, telehealth is no doubt the way to go. But it is really important that we continue to look into these disparities, what's causing them, and how we can find a path forward,” said

Deborah Hartzfeld, MS, CGC, of the Genomic Medicine Service based in Salt Lake City, Utah. She spoke in a presentation at the 2021 annual meeting of the Association of VA Hematology/Oncology (AVAHO) that was held virtually and in person in Denver, Colorado, from September 24 to September 26, 2021.

 

As Harzfeld explained, the genetic counselor workforce is expanding along with the number of indications for genetic testing, especially in cancer, “where the need for germline genetic testing for inherited cancer genes becomes broader every year.”

 

Genetic counselors are a homogenous group, she said, as revealed by a 2021 survey of most of the nation’s 5,629 certified generic counselors. The North American survey, by the National Society of Genetic Counselors, found that 94% of respondents identified themselves as female, and 90% were white/non-Hispanic.  

 

The survey report also noted that “the genetic counseling profession has grown by over 100% in the last 10 years and is expected to grow another 100% over the next 10 years. By 2025 there should be nearly 7,500 certified genetic counselors, and by 2030 there are likely to be over 10,000.”

 

Genetic counseling within the VA has also grown rapidly. In 2010, Harzfeld said, about 737 veterans were referred for the service. In 2020, the number was about 10,000, with about half referred for personal or family history of cancer.

 

The VA has 18 genetic counselors, not all of whom are actively seeing patients or working full time, she said. “Per the National Society of Genetic Counselors, there's one clinical genetic counselor per 100,000 people in the general population,” she said. “It's one for about 474,000 in the VA.”

 

Wait times for genetic counseling within the VA exceed Mission Act standards outside of urgent referrals in matters such as surgical or medical management, she said. “We usually see those patients within a week, but other folks have to wait or are referred into the community. It remains unclear how many of our patients could access care easily in the community or what the wait times at any individual VA will be.”

 

Fortunately, she said, telemedicine has increased access to genetic counseling within the general population and the VA, Harzfeld said. “A recent systematic evidence review found providing genetic counseling via video or telephone is comparable to in-person care, it increases access and it's likely feasible and acceptable to major stakeholders. It's worth noting that the data in this evidence review was collected prior to COVID-19 when fewer programs were using telehealth.”

 

Genetic counseling works especially well via telehealth because counselors don’t perform physical examinations, she said. “Prior to COVID, service probably saw maybe 4 VVC [VA Video Connect] appointments per month for genetic counseling. Now, VVC makes up about 70% of our new patient encounters. About 25% are telephone and about 5% are clinical video telehealth where the veteran goes into their clinic to be seated in front of the machine.”

 

Research has suggested that non-White patients are 40 to 50% less likely to be referred to telehealth for genetic counseling vs. in-person encounters, she said, although women in general (including black women) are more likely to be referred.

 

Harzfeld highlighted several challenges facing genetic counseling in the VA. She notes that contracted laboratories aren’t “really set up to be experts in germline genetic testing, so they’re not as nimble, and their test catalogs are not most likely going to be as comprehensive enough for what is needed.” Also, she said, “test ordering can be quite burdensome.”

 

“We need to continue working with various partners to increase access and the ease of ordering genetic testing,” she said.

 

Hartzfeld reports no disclosures.

 

The U.S. Department of Veteran Affairs (VA) has been unable to provide genetic counseling to veterans at the same level as the civilian community, and other gaps exist, a genetic counselor told oncologist and hematologist colleagues. The good news is that telemedicine is turning out to be a valuable and proven way to reach veterans who need this kind of care, she said, although certain patients are being left behind.

 

“To me, telehealth is no doubt the way to go. But it is really important that we continue to look into these disparities, what's causing them, and how we can find a path forward,” said

Deborah Hartzfeld, MS, CGC, of the Genomic Medicine Service based in Salt Lake City, Utah. She spoke in a presentation at the 2021 annual meeting of the Association of VA Hematology/Oncology (AVAHO) that was held virtually and in person in Denver, Colorado, from September 24 to September 26, 2021.

 

As Harzfeld explained, the genetic counselor workforce is expanding along with the number of indications for genetic testing, especially in cancer, “where the need for germline genetic testing for inherited cancer genes becomes broader every year.”

 

Genetic counselors are a homogenous group, she said, as revealed by a 2021 survey of most of the nation’s 5,629 certified generic counselors. The North American survey, by the National Society of Genetic Counselors, found that 94% of respondents identified themselves as female, and 90% were white/non-Hispanic.  

 

The survey report also noted that “the genetic counseling profession has grown by over 100% in the last 10 years and is expected to grow another 100% over the next 10 years. By 2025 there should be nearly 7,500 certified genetic counselors, and by 2030 there are likely to be over 10,000.”

 

Genetic counseling within the VA has also grown rapidly. In 2010, Harzfeld said, about 737 veterans were referred for the service. In 2020, the number was about 10,000, with about half referred for personal or family history of cancer.

 

The VA has 18 genetic counselors, not all of whom are actively seeing patients or working full time, she said. “Per the National Society of Genetic Counselors, there's one clinical genetic counselor per 100,000 people in the general population,” she said. “It's one for about 474,000 in the VA.”

 

Wait times for genetic counseling within the VA exceed Mission Act standards outside of urgent referrals in matters such as surgical or medical management, she said. “We usually see those patients within a week, but other folks have to wait or are referred into the community. It remains unclear how many of our patients could access care easily in the community or what the wait times at any individual VA will be.”

 

Fortunately, she said, telemedicine has increased access to genetic counseling within the general population and the VA, Harzfeld said. “A recent systematic evidence review found providing genetic counseling via video or telephone is comparable to in-person care, it increases access and it's likely feasible and acceptable to major stakeholders. It's worth noting that the data in this evidence review was collected prior to COVID-19 when fewer programs were using telehealth.”

 

Genetic counseling works especially well via telehealth because counselors don’t perform physical examinations, she said. “Prior to COVID, service probably saw maybe 4 VVC [VA Video Connect] appointments per month for genetic counseling. Now, VVC makes up about 70% of our new patient encounters. About 25% are telephone and about 5% are clinical video telehealth where the veteran goes into their clinic to be seated in front of the machine.”

 

Research has suggested that non-White patients are 40 to 50% less likely to be referred to telehealth for genetic counseling vs. in-person encounters, she said, although women in general (including black women) are more likely to be referred.

 

Harzfeld highlighted several challenges facing genetic counseling in the VA. She notes that contracted laboratories aren’t “really set up to be experts in germline genetic testing, so they’re not as nimble, and their test catalogs are not most likely going to be as comprehensive enough for what is needed.” Also, she said, “test ordering can be quite burdensome.”

 

“We need to continue working with various partners to increase access and the ease of ordering genetic testing,” she said.

 

Hartzfeld reports no disclosures.

 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 10/01/2021 - 12:00
Un-Gate On Date
Fri, 10/01/2021 - 12:00
Use ProPublica
CFC Schedule Remove Status
Fri, 10/01/2021 - 12:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

COVID boosters help protect blood cancer patients, but some still vulnerable

Article Type
Changed
Fri, 12/16/2022 - 11:59

Patients with hematologic malignancies who are seronegative after full SARS-CoV-2 vaccination may benefit from a booster, but breakthrough infections will remain a risk, particularly in those with B-cell malignancies, an observational study suggests.

Dr. Gwen Nichols

The findings, based on a review of COVID-19 booster vaccine recipients with B cell–derived hematologic malignancies from the prospective Leukemia & Lymphoma Society National Registry, provide valuable information about booster vaccinations in this vulnerable population, according to LLS chief medical officer and lead study author Gwen Nichols, MD.
 

The LLS Registry data

Of 49 patients included in the review, 38 failed to make antispike (anti-S) antibodies after full vaccination, and of those, 21 seroconverted after booster vaccination and 11 experienced seroelevation after the booster.

The patients who did not seroconvert were seronegative after initial vaccination and experienced no change in antibody level after the booster vaccination. In contrast, the 21 who seroconverted had a median 23.1 AU/ml increase in antibody level, Dr. Nichols, along with LLS chief scientific officer Lee M. Greenberger, PhD,and their colleagues reported online Sept. 6, 2021, in Cancer Cell.

Similar proportions of seroconverted patients had low-level responses between 2.2 and 23.1 AU/mL and robust response between 125 and 2,500 AU/mL, they noted. The 11 patients with seroelevation were seropositive after full vaccination and demonstrated a median increase of 2,128 AU/mL in antibody level after the booster vaccination.
 

Therapy effects on vaccine response

Outcomes of the current analysis also confirmed the authors’ previous finding, which suggested that “both disease and therapies can affect the serological response to vaccination,” they wrote, explaining that, among the 12 patients who received no malignancy-targeted treatments in the past 2 years, only 1 was a nonresponder, 7 demonstrated seroconversion, and 4 demonstrated seroelevation.

“In contrast, among the 21 patients who completed therapy with anti-CD20 antibodies either alone or in combination with other therapies, 12 patients were nonresponders, 7 patients demonstrated seroconversion, and 2 patients demonstrated seroelevation,” they added.

The authors also noted that five of seven patients who completed anti-CD20 antibody therapy alone or in combination with chemotherapy at least 7 months prior to the booster vaccination demonstrated seroconversion or seroelevation, whereas many of the patients with recent or maintenance anti-CD20 antibody therapy before the booster vaccination failed to seroconvert afterward.

In light of previous findings showing B-cell recovery begins 6-9 months after rituximab therapy, these data suggest that recent anti-CD20 antibody-containing treatment regimens may suppress booster vaccination response, the authors wrote.

The current data also support the group’s prior finding that use of a Bruton tyrosine kinase inhibitor may suppress vaccine response: Of the patients in the current study who experienced seroelevation and were treated with a BTKi, two discontinued BTKi therapy 7-23 months prior to booster vaccination, one maintained a low dose of ibrutinib before booster vaccination, one maintained BTKi therapy continuously before and after the booster, and the two who experienced marked seroconversion after booster vaccination stopped BTKi therapy at least 4 months prior.

Conversely, five patients with a very weak seroconversion and two patients with moderate seroconversion maintained BTKi therapy during booster vaccination.

“These data suggest that BTKi therapy can interfere with a response to booster vaccination,” they wrote, noting, however, that “it is encouraging that seven patients ... maintained on a BTKi seroconverted or experienced seroelevation after booster vaccination and [this] is consistent with a previous report on one patient.”
 

 

 

Study limitations and lessons

Although the findings of this study are limited by the small number of patients, the fact that treatment and disease were patient reported, and a lack of data on “antibody responses, particularly to the delta variant, B-cell memory, or T-cell responses,” they nevertheless provide encouraging news, Dr. Nichols told this news organization.

“Many blood cancer patients are getting boosters and a good number are able to make antibody with an additional dose. This is giving us much needed information about boosters,” she said. “Through the LLS National Patient Registry, we anticipate having data on hundreds of more patients over the course of the next few months.”

The information is needed because data suggest that up to 25% of patients with hematologic malignancies fail to make anti-S antibodies after full COVID-19 vaccination and that seronegative patients may be especially vulnerable to breakthrough infections, she and her colleagues noted.

Patients with B-cell malignancies are at the highest risk, and this is particularly concerning as some patients with blood cancer who contracted COVID-19 in the prevaccine period of the pandemic had “prolonged, severe infections; generated variant strains; and demonstrated significantly higher mortality rates compared to the general population,” they said.

However, a recently published placebo-controlled trial that demonstrated a booster vaccination–mediated increase in anti-S antibodies and neutralizing antibodies in immunosuppressed patients, and the current review, which focused on patients who obtained booster vaccinations prior to Aug. 12, 2021 (when the Food and Drug Administration granted emergency use authorization for booster doses in immunocompromised people), offer findings that suggest these patients may benefit from receiving COVID-19 boosters.

“We conclude that some patients with hematologic malignancies who are seronegative after a full course of vaccination may benefit from a booster,” the authors wrote. They added a warning: “Regulators, patients, and health care providers should be aware that a sizable subset of patients with blood cancer may remain at risk of breakthrough COVID-19 infections after full vaccination followed by booster vaccination.”

Dr. Nichols stressed that the findings “do not in any way suggest that blood cancer patients should stop therapy to get an antibody response to the vaccinations.”

“LLS is encouraging blood cancer patients to get vaccinated and to continue taking preventive measures such as wearing masks, social distancing, hand washing, and avoiding crowds and poorly ventilated indoor spaces,” she said.

Sergio Giralt, MD, professor and deputy head of the division of hematologic malignancies at Memorial Sloan Kettering Cancer Center, New York, further emphasized the importance of preventive measures.

“I think the extra booster will go a long way to protect our patients at this time but should not be used as a replacement for masking indoors and continued social distancing in this vulnerable patient population,” he said.

This study was supported by the LLS.

Publications
Topics
Sections

Patients with hematologic malignancies who are seronegative after full SARS-CoV-2 vaccination may benefit from a booster, but breakthrough infections will remain a risk, particularly in those with B-cell malignancies, an observational study suggests.

Dr. Gwen Nichols

The findings, based on a review of COVID-19 booster vaccine recipients with B cell–derived hematologic malignancies from the prospective Leukemia & Lymphoma Society National Registry, provide valuable information about booster vaccinations in this vulnerable population, according to LLS chief medical officer and lead study author Gwen Nichols, MD.
 

The LLS Registry data

Of 49 patients included in the review, 38 failed to make antispike (anti-S) antibodies after full vaccination, and of those, 21 seroconverted after booster vaccination and 11 experienced seroelevation after the booster.

The patients who did not seroconvert were seronegative after initial vaccination and experienced no change in antibody level after the booster vaccination. In contrast, the 21 who seroconverted had a median 23.1 AU/ml increase in antibody level, Dr. Nichols, along with LLS chief scientific officer Lee M. Greenberger, PhD,and their colleagues reported online Sept. 6, 2021, in Cancer Cell.

Similar proportions of seroconverted patients had low-level responses between 2.2 and 23.1 AU/mL and robust response between 125 and 2,500 AU/mL, they noted. The 11 patients with seroelevation were seropositive after full vaccination and demonstrated a median increase of 2,128 AU/mL in antibody level after the booster vaccination.
 

Therapy effects on vaccine response

Outcomes of the current analysis also confirmed the authors’ previous finding, which suggested that “both disease and therapies can affect the serological response to vaccination,” they wrote, explaining that, among the 12 patients who received no malignancy-targeted treatments in the past 2 years, only 1 was a nonresponder, 7 demonstrated seroconversion, and 4 demonstrated seroelevation.

“In contrast, among the 21 patients who completed therapy with anti-CD20 antibodies either alone or in combination with other therapies, 12 patients were nonresponders, 7 patients demonstrated seroconversion, and 2 patients demonstrated seroelevation,” they added.

The authors also noted that five of seven patients who completed anti-CD20 antibody therapy alone or in combination with chemotherapy at least 7 months prior to the booster vaccination demonstrated seroconversion or seroelevation, whereas many of the patients with recent or maintenance anti-CD20 antibody therapy before the booster vaccination failed to seroconvert afterward.

In light of previous findings showing B-cell recovery begins 6-9 months after rituximab therapy, these data suggest that recent anti-CD20 antibody-containing treatment regimens may suppress booster vaccination response, the authors wrote.

The current data also support the group’s prior finding that use of a Bruton tyrosine kinase inhibitor may suppress vaccine response: Of the patients in the current study who experienced seroelevation and were treated with a BTKi, two discontinued BTKi therapy 7-23 months prior to booster vaccination, one maintained a low dose of ibrutinib before booster vaccination, one maintained BTKi therapy continuously before and after the booster, and the two who experienced marked seroconversion after booster vaccination stopped BTKi therapy at least 4 months prior.

Conversely, five patients with a very weak seroconversion and two patients with moderate seroconversion maintained BTKi therapy during booster vaccination.

“These data suggest that BTKi therapy can interfere with a response to booster vaccination,” they wrote, noting, however, that “it is encouraging that seven patients ... maintained on a BTKi seroconverted or experienced seroelevation after booster vaccination and [this] is consistent with a previous report on one patient.”
 

 

 

Study limitations and lessons

Although the findings of this study are limited by the small number of patients, the fact that treatment and disease were patient reported, and a lack of data on “antibody responses, particularly to the delta variant, B-cell memory, or T-cell responses,” they nevertheless provide encouraging news, Dr. Nichols told this news organization.

“Many blood cancer patients are getting boosters and a good number are able to make antibody with an additional dose. This is giving us much needed information about boosters,” she said. “Through the LLS National Patient Registry, we anticipate having data on hundreds of more patients over the course of the next few months.”

The information is needed because data suggest that up to 25% of patients with hematologic malignancies fail to make anti-S antibodies after full COVID-19 vaccination and that seronegative patients may be especially vulnerable to breakthrough infections, she and her colleagues noted.

Patients with B-cell malignancies are at the highest risk, and this is particularly concerning as some patients with blood cancer who contracted COVID-19 in the prevaccine period of the pandemic had “prolonged, severe infections; generated variant strains; and demonstrated significantly higher mortality rates compared to the general population,” they said.

However, a recently published placebo-controlled trial that demonstrated a booster vaccination–mediated increase in anti-S antibodies and neutralizing antibodies in immunosuppressed patients, and the current review, which focused on patients who obtained booster vaccinations prior to Aug. 12, 2021 (when the Food and Drug Administration granted emergency use authorization for booster doses in immunocompromised people), offer findings that suggest these patients may benefit from receiving COVID-19 boosters.

“We conclude that some patients with hematologic malignancies who are seronegative after a full course of vaccination may benefit from a booster,” the authors wrote. They added a warning: “Regulators, patients, and health care providers should be aware that a sizable subset of patients with blood cancer may remain at risk of breakthrough COVID-19 infections after full vaccination followed by booster vaccination.”

Dr. Nichols stressed that the findings “do not in any way suggest that blood cancer patients should stop therapy to get an antibody response to the vaccinations.”

“LLS is encouraging blood cancer patients to get vaccinated and to continue taking preventive measures such as wearing masks, social distancing, hand washing, and avoiding crowds and poorly ventilated indoor spaces,” she said.

Sergio Giralt, MD, professor and deputy head of the division of hematologic malignancies at Memorial Sloan Kettering Cancer Center, New York, further emphasized the importance of preventive measures.

“I think the extra booster will go a long way to protect our patients at this time but should not be used as a replacement for masking indoors and continued social distancing in this vulnerable patient population,” he said.

This study was supported by the LLS.

Patients with hematologic malignancies who are seronegative after full SARS-CoV-2 vaccination may benefit from a booster, but breakthrough infections will remain a risk, particularly in those with B-cell malignancies, an observational study suggests.

Dr. Gwen Nichols

The findings, based on a review of COVID-19 booster vaccine recipients with B cell–derived hematologic malignancies from the prospective Leukemia & Lymphoma Society National Registry, provide valuable information about booster vaccinations in this vulnerable population, according to LLS chief medical officer and lead study author Gwen Nichols, MD.
 

The LLS Registry data

Of 49 patients included in the review, 38 failed to make antispike (anti-S) antibodies after full vaccination, and of those, 21 seroconverted after booster vaccination and 11 experienced seroelevation after the booster.

The patients who did not seroconvert were seronegative after initial vaccination and experienced no change in antibody level after the booster vaccination. In contrast, the 21 who seroconverted had a median 23.1 AU/ml increase in antibody level, Dr. Nichols, along with LLS chief scientific officer Lee M. Greenberger, PhD,and their colleagues reported online Sept. 6, 2021, in Cancer Cell.

Similar proportions of seroconverted patients had low-level responses between 2.2 and 23.1 AU/mL and robust response between 125 and 2,500 AU/mL, they noted. The 11 patients with seroelevation were seropositive after full vaccination and demonstrated a median increase of 2,128 AU/mL in antibody level after the booster vaccination.
 

Therapy effects on vaccine response

Outcomes of the current analysis also confirmed the authors’ previous finding, which suggested that “both disease and therapies can affect the serological response to vaccination,” they wrote, explaining that, among the 12 patients who received no malignancy-targeted treatments in the past 2 years, only 1 was a nonresponder, 7 demonstrated seroconversion, and 4 demonstrated seroelevation.

“In contrast, among the 21 patients who completed therapy with anti-CD20 antibodies either alone or in combination with other therapies, 12 patients were nonresponders, 7 patients demonstrated seroconversion, and 2 patients demonstrated seroelevation,” they added.

The authors also noted that five of seven patients who completed anti-CD20 antibody therapy alone or in combination with chemotherapy at least 7 months prior to the booster vaccination demonstrated seroconversion or seroelevation, whereas many of the patients with recent or maintenance anti-CD20 antibody therapy before the booster vaccination failed to seroconvert afterward.

In light of previous findings showing B-cell recovery begins 6-9 months after rituximab therapy, these data suggest that recent anti-CD20 antibody-containing treatment regimens may suppress booster vaccination response, the authors wrote.

The current data also support the group’s prior finding that use of a Bruton tyrosine kinase inhibitor may suppress vaccine response: Of the patients in the current study who experienced seroelevation and were treated with a BTKi, two discontinued BTKi therapy 7-23 months prior to booster vaccination, one maintained a low dose of ibrutinib before booster vaccination, one maintained BTKi therapy continuously before and after the booster, and the two who experienced marked seroconversion after booster vaccination stopped BTKi therapy at least 4 months prior.

Conversely, five patients with a very weak seroconversion and two patients with moderate seroconversion maintained BTKi therapy during booster vaccination.

“These data suggest that BTKi therapy can interfere with a response to booster vaccination,” they wrote, noting, however, that “it is encouraging that seven patients ... maintained on a BTKi seroconverted or experienced seroelevation after booster vaccination and [this] is consistent with a previous report on one patient.”
 

 

 

Study limitations and lessons

Although the findings of this study are limited by the small number of patients, the fact that treatment and disease were patient reported, and a lack of data on “antibody responses, particularly to the delta variant, B-cell memory, or T-cell responses,” they nevertheless provide encouraging news, Dr. Nichols told this news organization.

“Many blood cancer patients are getting boosters and a good number are able to make antibody with an additional dose. This is giving us much needed information about boosters,” she said. “Through the LLS National Patient Registry, we anticipate having data on hundreds of more patients over the course of the next few months.”

The information is needed because data suggest that up to 25% of patients with hematologic malignancies fail to make anti-S antibodies after full COVID-19 vaccination and that seronegative patients may be especially vulnerable to breakthrough infections, she and her colleagues noted.

Patients with B-cell malignancies are at the highest risk, and this is particularly concerning as some patients with blood cancer who contracted COVID-19 in the prevaccine period of the pandemic had “prolonged, severe infections; generated variant strains; and demonstrated significantly higher mortality rates compared to the general population,” they said.

However, a recently published placebo-controlled trial that demonstrated a booster vaccination–mediated increase in anti-S antibodies and neutralizing antibodies in immunosuppressed patients, and the current review, which focused on patients who obtained booster vaccinations prior to Aug. 12, 2021 (when the Food and Drug Administration granted emergency use authorization for booster doses in immunocompromised people), offer findings that suggest these patients may benefit from receiving COVID-19 boosters.

“We conclude that some patients with hematologic malignancies who are seronegative after a full course of vaccination may benefit from a booster,” the authors wrote. They added a warning: “Regulators, patients, and health care providers should be aware that a sizable subset of patients with blood cancer may remain at risk of breakthrough COVID-19 infections after full vaccination followed by booster vaccination.”

Dr. Nichols stressed that the findings “do not in any way suggest that blood cancer patients should stop therapy to get an antibody response to the vaccinations.”

“LLS is encouraging blood cancer patients to get vaccinated and to continue taking preventive measures such as wearing masks, social distancing, hand washing, and avoiding crowds and poorly ventilated indoor spaces,” she said.

Sergio Giralt, MD, professor and deputy head of the division of hematologic malignancies at Memorial Sloan Kettering Cancer Center, New York, further emphasized the importance of preventive measures.

“I think the extra booster will go a long way to protect our patients at this time but should not be used as a replacement for masking indoors and continued social distancing in this vulnerable patient population,” he said.

This study was supported by the LLS.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CANCER CELL

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

FDA okays new oral CGRP antagonist for migraine prevention

Article Type
Changed
Thu, 12/15/2022 - 14:36

The Food and Drug Administration has approved atogepant (Qulipta), a novel calcitonin gene-related peptide (CGRP) receptor antagonist, for the prevention of episodic migraine, the manufacturer announced in a release.

The once-daily medication will be available in doses of 10 mg, 30 mg, and 60 mg.

“Qulipta provides a simple oral treatment option specifically developed to prevent migraine attacks and target CGRP, which is believed to be crucially involved in migraine in many patients,” coinvestigator Peter J. Goadsby, MD, PhD, DSc, neurologist and professor at the University of California, Los Angeles, and King’s College London, said in the release.

Approval was based partly on the findings from the phase 3 ADVANCE trial, in which patients with episodic migraine were randomly assigned to receive placebo or a 10-mg, 30-mg, or 60-mg daily dose of atogepant for 12 weeks.

As reported by this news organization, all three doses of atogepant reduced the number of mean monthly migraine days.

With this approval, neurologists will be able to choose from four monoclonal antibodies and two gepants for the preventive treatment of migraine.

“Having another gepant that can also be given preventively is a good idea, because one may be better than the other for a patient,” Alan M. Rapoport, MD, past president of the International Headache Society and founder and director emeritus of the New England Center for Headache, Stamford, Conn., told this news organization.

“Once we have a year or so of experience with atogepant, we’ll have a pretty good idea of which one works better preventively,” said Dr. Rapoport, who was not involved with the research.
 

Practice changing?

In the ADVANCE trial, there was a reduction of 3.69 migraine days with the 10-mg dose, 3.86 days with the 30-mg dose, and 4.2 days with the 60-mg dose. Placebo was associated with a reduction of 2.48 migraine days.

In addition, more than half of patients in each atogepant arm achieved a reduction in mean monthly migraine days of 50% or greater. This outcome occurred in 55.6% of the 10-mg atogepant group, 58.7% of the 30-mg group, and 60.8% of the 60-mg group. Approximately 29% patients who received placebo achieved this outcome.

The data indicated that atogepant has a favorable safety profile. The most common adverse events associated with treatment were constipation, nausea, and upper respiratory tract infection.

Dr. Rapoport, who is also a clinical professor of neurology at UCLA, noted that he was impressed with the efficacy.

“I’m not as impressed with the adverse events, but they’re not serious, and they don’t necessarily last,” he said.

Although being able to prescribe a single drug for acute and preventive treatment may be an advantage, it remains to be seen whether the tolerability and price of atogepant will be barriers for patients, Dr. Rapoport added.

How the approval will affect clinical practice is also unclear, he noted.

“If you’re going to start someone on a preventive, especially if it’s a woman of childbearing potential, you might just consider one of the two gepants. Doctors will decide once they see how they work,” said Dr. Rapoport.
 

 

 

Not a ‘breakthrough’ treatment

Also commenting ahead of the approval, Elizabeth W. Loder, MD, vice chair for academic affairs in the department of neurology at Brigham and Women’s Hospital, Boston, noted that the “safety of these CGRP medications in pregnancy is uncertain, and there are theoretical reasons to be concerned about it.”

Unlike injectable CGRP medications, atogepant is eliminated from the body relatively quickly after the patient stops taking it, said Dr. Loder, who is also professor of neurology at Harvard Medical School, Boston. However, atogepant may not otherwise differ greatly from other medications of its type.

“I don’t see a reason to think that one of these oral CGRP medicines is much more effective than another one,” said Dr. Loder.

“In my mind, as a clinician who will be prescribing these for patients, it will be cost and the ease of getting it covered that makes the difference,” she added.

These questions may raise concerns. “Those of us who treat patients who do not have private insurance find it very difficult to get these medications for them, even in situations where they have exhausted other alternatives,” said Dr. Loder.

Patients insured by Medicare or Medicaid “usually have no avenue to get some of these new, expensive treatments,” she said.

The approval of atogepant for acute and preventive treatment shows that the distinction between these indications may be artificial, Dr. Loder noted. The approval “will, I hope, help people think more flexibly about the way in which we use medications.”

It is a positive that atogepant has emerged as another option for preventive therapy, but the treatment cannot be considered a breakthrough, Dr. Loder added. The efficacy of atogepant, like that of other preventive treatments for migraine, is modest.

“It would be so nice if we could find things that were more effective than the treatments we currently have,” said Dr. Loder.

A version of this article first appeared on Medscape.com.

Issue
Neurology Reviews - 29(11)
Publications
Topics
Sections

The Food and Drug Administration has approved atogepant (Qulipta), a novel calcitonin gene-related peptide (CGRP) receptor antagonist, for the prevention of episodic migraine, the manufacturer announced in a release.

The once-daily medication will be available in doses of 10 mg, 30 mg, and 60 mg.

“Qulipta provides a simple oral treatment option specifically developed to prevent migraine attacks and target CGRP, which is believed to be crucially involved in migraine in many patients,” coinvestigator Peter J. Goadsby, MD, PhD, DSc, neurologist and professor at the University of California, Los Angeles, and King’s College London, said in the release.

Approval was based partly on the findings from the phase 3 ADVANCE trial, in which patients with episodic migraine were randomly assigned to receive placebo or a 10-mg, 30-mg, or 60-mg daily dose of atogepant for 12 weeks.

As reported by this news organization, all three doses of atogepant reduced the number of mean monthly migraine days.

With this approval, neurologists will be able to choose from four monoclonal antibodies and two gepants for the preventive treatment of migraine.

“Having another gepant that can also be given preventively is a good idea, because one may be better than the other for a patient,” Alan M. Rapoport, MD, past president of the International Headache Society and founder and director emeritus of the New England Center for Headache, Stamford, Conn., told this news organization.

“Once we have a year or so of experience with atogepant, we’ll have a pretty good idea of which one works better preventively,” said Dr. Rapoport, who was not involved with the research.
 

Practice changing?

In the ADVANCE trial, there was a reduction of 3.69 migraine days with the 10-mg dose, 3.86 days with the 30-mg dose, and 4.2 days with the 60-mg dose. Placebo was associated with a reduction of 2.48 migraine days.

In addition, more than half of patients in each atogepant arm achieved a reduction in mean monthly migraine days of 50% or greater. This outcome occurred in 55.6% of the 10-mg atogepant group, 58.7% of the 30-mg group, and 60.8% of the 60-mg group. Approximately 29% patients who received placebo achieved this outcome.

The data indicated that atogepant has a favorable safety profile. The most common adverse events associated with treatment were constipation, nausea, and upper respiratory tract infection.

Dr. Rapoport, who is also a clinical professor of neurology at UCLA, noted that he was impressed with the efficacy.

“I’m not as impressed with the adverse events, but they’re not serious, and they don’t necessarily last,” he said.

Although being able to prescribe a single drug for acute and preventive treatment may be an advantage, it remains to be seen whether the tolerability and price of atogepant will be barriers for patients, Dr. Rapoport added.

How the approval will affect clinical practice is also unclear, he noted.

“If you’re going to start someone on a preventive, especially if it’s a woman of childbearing potential, you might just consider one of the two gepants. Doctors will decide once they see how they work,” said Dr. Rapoport.
 

 

 

Not a ‘breakthrough’ treatment

Also commenting ahead of the approval, Elizabeth W. Loder, MD, vice chair for academic affairs in the department of neurology at Brigham and Women’s Hospital, Boston, noted that the “safety of these CGRP medications in pregnancy is uncertain, and there are theoretical reasons to be concerned about it.”

Unlike injectable CGRP medications, atogepant is eliminated from the body relatively quickly after the patient stops taking it, said Dr. Loder, who is also professor of neurology at Harvard Medical School, Boston. However, atogepant may not otherwise differ greatly from other medications of its type.

“I don’t see a reason to think that one of these oral CGRP medicines is much more effective than another one,” said Dr. Loder.

“In my mind, as a clinician who will be prescribing these for patients, it will be cost and the ease of getting it covered that makes the difference,” she added.

These questions may raise concerns. “Those of us who treat patients who do not have private insurance find it very difficult to get these medications for them, even in situations where they have exhausted other alternatives,” said Dr. Loder.

Patients insured by Medicare or Medicaid “usually have no avenue to get some of these new, expensive treatments,” she said.

The approval of atogepant for acute and preventive treatment shows that the distinction between these indications may be artificial, Dr. Loder noted. The approval “will, I hope, help people think more flexibly about the way in which we use medications.”

It is a positive that atogepant has emerged as another option for preventive therapy, but the treatment cannot be considered a breakthrough, Dr. Loder added. The efficacy of atogepant, like that of other preventive treatments for migraine, is modest.

“It would be so nice if we could find things that were more effective than the treatments we currently have,” said Dr. Loder.

A version of this article first appeared on Medscape.com.

The Food and Drug Administration has approved atogepant (Qulipta), a novel calcitonin gene-related peptide (CGRP) receptor antagonist, for the prevention of episodic migraine, the manufacturer announced in a release.

The once-daily medication will be available in doses of 10 mg, 30 mg, and 60 mg.

“Qulipta provides a simple oral treatment option specifically developed to prevent migraine attacks and target CGRP, which is believed to be crucially involved in migraine in many patients,” coinvestigator Peter J. Goadsby, MD, PhD, DSc, neurologist and professor at the University of California, Los Angeles, and King’s College London, said in the release.

Approval was based partly on the findings from the phase 3 ADVANCE trial, in which patients with episodic migraine were randomly assigned to receive placebo or a 10-mg, 30-mg, or 60-mg daily dose of atogepant for 12 weeks.

As reported by this news organization, all three doses of atogepant reduced the number of mean monthly migraine days.

With this approval, neurologists will be able to choose from four monoclonal antibodies and two gepants for the preventive treatment of migraine.

“Having another gepant that can also be given preventively is a good idea, because one may be better than the other for a patient,” Alan M. Rapoport, MD, past president of the International Headache Society and founder and director emeritus of the New England Center for Headache, Stamford, Conn., told this news organization.

“Once we have a year or so of experience with atogepant, we’ll have a pretty good idea of which one works better preventively,” said Dr. Rapoport, who was not involved with the research.
 

Practice changing?

In the ADVANCE trial, there was a reduction of 3.69 migraine days with the 10-mg dose, 3.86 days with the 30-mg dose, and 4.2 days with the 60-mg dose. Placebo was associated with a reduction of 2.48 migraine days.

In addition, more than half of patients in each atogepant arm achieved a reduction in mean monthly migraine days of 50% or greater. This outcome occurred in 55.6% of the 10-mg atogepant group, 58.7% of the 30-mg group, and 60.8% of the 60-mg group. Approximately 29% patients who received placebo achieved this outcome.

The data indicated that atogepant has a favorable safety profile. The most common adverse events associated with treatment were constipation, nausea, and upper respiratory tract infection.

Dr. Rapoport, who is also a clinical professor of neurology at UCLA, noted that he was impressed with the efficacy.

“I’m not as impressed with the adverse events, but they’re not serious, and they don’t necessarily last,” he said.

Although being able to prescribe a single drug for acute and preventive treatment may be an advantage, it remains to be seen whether the tolerability and price of atogepant will be barriers for patients, Dr. Rapoport added.

How the approval will affect clinical practice is also unclear, he noted.

“If you’re going to start someone on a preventive, especially if it’s a woman of childbearing potential, you might just consider one of the two gepants. Doctors will decide once they see how they work,” said Dr. Rapoport.
 

 

 

Not a ‘breakthrough’ treatment

Also commenting ahead of the approval, Elizabeth W. Loder, MD, vice chair for academic affairs in the department of neurology at Brigham and Women’s Hospital, Boston, noted that the “safety of these CGRP medications in pregnancy is uncertain, and there are theoretical reasons to be concerned about it.”

Unlike injectable CGRP medications, atogepant is eliminated from the body relatively quickly after the patient stops taking it, said Dr. Loder, who is also professor of neurology at Harvard Medical School, Boston. However, atogepant may not otherwise differ greatly from other medications of its type.

“I don’t see a reason to think that one of these oral CGRP medicines is much more effective than another one,” said Dr. Loder.

“In my mind, as a clinician who will be prescribing these for patients, it will be cost and the ease of getting it covered that makes the difference,” she added.

These questions may raise concerns. “Those of us who treat patients who do not have private insurance find it very difficult to get these medications for them, even in situations where they have exhausted other alternatives,” said Dr. Loder.

Patients insured by Medicare or Medicaid “usually have no avenue to get some of these new, expensive treatments,” she said.

The approval of atogepant for acute and preventive treatment shows that the distinction between these indications may be artificial, Dr. Loder noted. The approval “will, I hope, help people think more flexibly about the way in which we use medications.”

It is a positive that atogepant has emerged as another option for preventive therapy, but the treatment cannot be considered a breakthrough, Dr. Loder added. The efficacy of atogepant, like that of other preventive treatments for migraine, is modest.

“It would be so nice if we could find things that were more effective than the treatments we currently have,” said Dr. Loder.

A version of this article first appeared on Medscape.com.

Issue
Neurology Reviews - 29(11)
Issue
Neurology Reviews - 29(11)
Publications
Publications
Topics
Article Type
Sections
Citation Override
Publish date: September 29, 2021
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

‘Locker room talk’ about death: Time for oncologists to stop

Article Type
Changed
Thu, 12/15/2022 - 14:36

In a recent inpatient service block, I was seeing patients alongside a resident I had gotten to know well. We were consulted on a patient with metastatic head and neck cancer who had not sought care for over a year.

When the patient presented, his voice was raspy and he could not swallow. He had lost 40 pounds. In addition to his locally advanced disease, his lungs were riddled with metastatic lesions.

When we left the room, the resident and I went to speak to the patient’s primary team, and he began to relay our recommendations.

The first words out of his mouth were, “Well, it’s pretty clear he’s going to die.”

The statement took me aback. I wasn’t alarmed by the accuracy of what he had said. The patient was obviously not doing well, and he ended up dying soon after this visit.

It was more the abrupt manner in which the resident had spoken about death. The brusque phrasing felt atypical coming from the otherwise gentle-hearted trainee. He wasn’t referring to a faceless person. We had just seen the man a few minutes ago and heard his personal struggles. I tried to see if anyone else on the team was caught off guard, but everyone was taking notes or continuing to listen, seemingly undeterred.
 

Oncologists’ ‘locker room talk’

I’ve noticed that “locker room talk” about death happens often. Phrases like “he’s definitely not going to do well” and “his life expectancy is poor” make their way into oncologists’ daily language. Thinking back on my own interactions, I realize I am also guilty of discussing death in this way.

And now, with the COVID pandemic forcing most of our tumor boards to go virtual, I find this locker room talk comes even more readily; phrases like “this patient is going to die” are often passed around flippantly, as if saying so will help ease the tension. During these interactions, my colleagues and I rarely acknowledge the seriousness of what a patient death will do to their family and loved ones – or what losing a patient whom we’ve known for years may do to our own psyche.

This language can even creep into how we speak with patients. We are often taught to offer prognoses coldly, ensuring that patients have a clear sense of how long they have left and to help inform their treatment choices. And yet, this training does not necessarily align with what patients want and need. For instance, in a recent survey of patients with chronic obstructive pulmonary disease, patients consistently rated physicians poorly at discussing prognosis, what dying might be like, as well as spirituality and religion.

But at the same time, these matter-of-fact statements about death probably help protect us. Death is a routine, inevitable part of an oncologist’s life, and over time, oncology training and practice hardens us to it. During medical school, I remember that a patient dying would trigger immediate reflection, sadness, and conversation with our peers. Now, unless I know a patient well, I find myself rarely reflecting on the patient behind the facts. This evolution is natural for an oncologist: If you don’t develop a tough skin about death, you may become overwhelmed with the frequency of it.

The COVID pandemic has amped our hardness toward death into overdrive. Whether we are in the intensive care unit or simply viewing death rates during the most recent COVID Delta wave, many of us cope by disassociating a face from a name.
 

 

 

Making time for reflection

But taking time to reflect can be therapeutic.

I recently referred a patient with metastatic prostate cancer for a phase 1 trial at an outside institution. He was one of the first patients in my genitourinary malignancies clinic when I started as an attending. The patient had progressed through several lines of therapy and was being referred for an investigational phase 1 therapy. We had discussed hospice referral, and the patient was ready for it if this therapy didn’t work out.

I did not see or hear from the man while he was on the trial. A few months later, however, the principal investigator of the trial called me to let me know the patient had progressed through the agent, suffering from significant urinary obstruction, and he was on hospice. “Unfortunately,” the investigator told me, “he’s not going to live much longer.”

When I checked in with the hospice, the patient had died.

I was surprised again at how matter-of-fact the discussion of death had been. But I was even more surprised by my own reaction. Despite the relationship I had formed with the patient, I did not feel much when I heard he had died. I didn’t have time to process the news in the moment. It was time to move on to the next patient.

It was only later, when I called the patient’s family, that I allowed my emotions to flood in. I told his family how grateful I was to know him, how strong he’d been. The patient’s family and I talked about the human, not his passing. It felt good.
 

Abandoning locker room talk

So how do we change how we talk about death? I don’t think the answer is massive educational programs or passing responsibility for advance care planning onto palliative care specialists. The change needs to be driven by individual oncologists. We can call out discussions of death that make us uncomfortable, gently reminding each other that we’re talking about a human life.

We can learn from our palliative care colleagues; their conversations about death routinely include a patient’s support system and personal stories. Palliative care doctors always refer to the patient by name, which helps humanize the person behind the chart.

We can emphasize a feeling of hope, a sentiment that may also be therapeutic to our patients. Even when a patient is dying, there is always something to be done. We can comfort their family, explaining what brought us to this point and how sorry we are that this is happening. We can provide options for symptom control and help patients manage those symptoms.

And we can allow ourselves to talk about how much a death affects us. We can acknowledge how much it sucks that a patient is going to die, how challenging that will be to his/her family, and how we wish it could have ended differently.

Subtle changes like these will improve our own ability to process and discuss death and will ultimately lead to better relationships with our patients. But it starts with eliminating the “locker room talk” of how we discuss death.
 

Ravi B. Parikh, MD, MPP, is a medical oncologist and faculty member at the University of Pennsylvania and the Philadelphia VA Medical Center, an adjunct fellow at the Leonard Davis Institute of Health Economics, and senior clinical advisor at the Coalition to Transform Advanced Care (C-TAC). He has served as a director, officer, partner, employee, adviser, consultant, or trustee for GNS Healthcare, Nanology, and Cancer Study Group, and he has received research grant from Embedded Healthcare, Veterans Administration, PCF, National Palliative Care Research Center, and MUSC. A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

In a recent inpatient service block, I was seeing patients alongside a resident I had gotten to know well. We were consulted on a patient with metastatic head and neck cancer who had not sought care for over a year.

When the patient presented, his voice was raspy and he could not swallow. He had lost 40 pounds. In addition to his locally advanced disease, his lungs were riddled with metastatic lesions.

When we left the room, the resident and I went to speak to the patient’s primary team, and he began to relay our recommendations.

The first words out of his mouth were, “Well, it’s pretty clear he’s going to die.”

The statement took me aback. I wasn’t alarmed by the accuracy of what he had said. The patient was obviously not doing well, and he ended up dying soon after this visit.

It was more the abrupt manner in which the resident had spoken about death. The brusque phrasing felt atypical coming from the otherwise gentle-hearted trainee. He wasn’t referring to a faceless person. We had just seen the man a few minutes ago and heard his personal struggles. I tried to see if anyone else on the team was caught off guard, but everyone was taking notes or continuing to listen, seemingly undeterred.
 

Oncologists’ ‘locker room talk’

I’ve noticed that “locker room talk” about death happens often. Phrases like “he’s definitely not going to do well” and “his life expectancy is poor” make their way into oncologists’ daily language. Thinking back on my own interactions, I realize I am also guilty of discussing death in this way.

And now, with the COVID pandemic forcing most of our tumor boards to go virtual, I find this locker room talk comes even more readily; phrases like “this patient is going to die” are often passed around flippantly, as if saying so will help ease the tension. During these interactions, my colleagues and I rarely acknowledge the seriousness of what a patient death will do to their family and loved ones – or what losing a patient whom we’ve known for years may do to our own psyche.

This language can even creep into how we speak with patients. We are often taught to offer prognoses coldly, ensuring that patients have a clear sense of how long they have left and to help inform their treatment choices. And yet, this training does not necessarily align with what patients want and need. For instance, in a recent survey of patients with chronic obstructive pulmonary disease, patients consistently rated physicians poorly at discussing prognosis, what dying might be like, as well as spirituality and religion.

But at the same time, these matter-of-fact statements about death probably help protect us. Death is a routine, inevitable part of an oncologist’s life, and over time, oncology training and practice hardens us to it. During medical school, I remember that a patient dying would trigger immediate reflection, sadness, and conversation with our peers. Now, unless I know a patient well, I find myself rarely reflecting on the patient behind the facts. This evolution is natural for an oncologist: If you don’t develop a tough skin about death, you may become overwhelmed with the frequency of it.

The COVID pandemic has amped our hardness toward death into overdrive. Whether we are in the intensive care unit or simply viewing death rates during the most recent COVID Delta wave, many of us cope by disassociating a face from a name.
 

 

 

Making time for reflection

But taking time to reflect can be therapeutic.

I recently referred a patient with metastatic prostate cancer for a phase 1 trial at an outside institution. He was one of the first patients in my genitourinary malignancies clinic when I started as an attending. The patient had progressed through several lines of therapy and was being referred for an investigational phase 1 therapy. We had discussed hospice referral, and the patient was ready for it if this therapy didn’t work out.

I did not see or hear from the man while he was on the trial. A few months later, however, the principal investigator of the trial called me to let me know the patient had progressed through the agent, suffering from significant urinary obstruction, and he was on hospice. “Unfortunately,” the investigator told me, “he’s not going to live much longer.”

When I checked in with the hospice, the patient had died.

I was surprised again at how matter-of-fact the discussion of death had been. But I was even more surprised by my own reaction. Despite the relationship I had formed with the patient, I did not feel much when I heard he had died. I didn’t have time to process the news in the moment. It was time to move on to the next patient.

It was only later, when I called the patient’s family, that I allowed my emotions to flood in. I told his family how grateful I was to know him, how strong he’d been. The patient’s family and I talked about the human, not his passing. It felt good.
 

Abandoning locker room talk

So how do we change how we talk about death? I don’t think the answer is massive educational programs or passing responsibility for advance care planning onto palliative care specialists. The change needs to be driven by individual oncologists. We can call out discussions of death that make us uncomfortable, gently reminding each other that we’re talking about a human life.

We can learn from our palliative care colleagues; their conversations about death routinely include a patient’s support system and personal stories. Palliative care doctors always refer to the patient by name, which helps humanize the person behind the chart.

We can emphasize a feeling of hope, a sentiment that may also be therapeutic to our patients. Even when a patient is dying, there is always something to be done. We can comfort their family, explaining what brought us to this point and how sorry we are that this is happening. We can provide options for symptom control and help patients manage those symptoms.

And we can allow ourselves to talk about how much a death affects us. We can acknowledge how much it sucks that a patient is going to die, how challenging that will be to his/her family, and how we wish it could have ended differently.

Subtle changes like these will improve our own ability to process and discuss death and will ultimately lead to better relationships with our patients. But it starts with eliminating the “locker room talk” of how we discuss death.
 

Ravi B. Parikh, MD, MPP, is a medical oncologist and faculty member at the University of Pennsylvania and the Philadelphia VA Medical Center, an adjunct fellow at the Leonard Davis Institute of Health Economics, and senior clinical advisor at the Coalition to Transform Advanced Care (C-TAC). He has served as a director, officer, partner, employee, adviser, consultant, or trustee for GNS Healthcare, Nanology, and Cancer Study Group, and he has received research grant from Embedded Healthcare, Veterans Administration, PCF, National Palliative Care Research Center, and MUSC. A version of this article first appeared on Medscape.com.

In a recent inpatient service block, I was seeing patients alongside a resident I had gotten to know well. We were consulted on a patient with metastatic head and neck cancer who had not sought care for over a year.

When the patient presented, his voice was raspy and he could not swallow. He had lost 40 pounds. In addition to his locally advanced disease, his lungs were riddled with metastatic lesions.

When we left the room, the resident and I went to speak to the patient’s primary team, and he began to relay our recommendations.

The first words out of his mouth were, “Well, it’s pretty clear he’s going to die.”

The statement took me aback. I wasn’t alarmed by the accuracy of what he had said. The patient was obviously not doing well, and he ended up dying soon after this visit.

It was more the abrupt manner in which the resident had spoken about death. The brusque phrasing felt atypical coming from the otherwise gentle-hearted trainee. He wasn’t referring to a faceless person. We had just seen the man a few minutes ago and heard his personal struggles. I tried to see if anyone else on the team was caught off guard, but everyone was taking notes or continuing to listen, seemingly undeterred.
 

Oncologists’ ‘locker room talk’

I’ve noticed that “locker room talk” about death happens often. Phrases like “he’s definitely not going to do well” and “his life expectancy is poor” make their way into oncologists’ daily language. Thinking back on my own interactions, I realize I am also guilty of discussing death in this way.

And now, with the COVID pandemic forcing most of our tumor boards to go virtual, I find this locker room talk comes even more readily; phrases like “this patient is going to die” are often passed around flippantly, as if saying so will help ease the tension. During these interactions, my colleagues and I rarely acknowledge the seriousness of what a patient death will do to their family and loved ones – or what losing a patient whom we’ve known for years may do to our own psyche.

This language can even creep into how we speak with patients. We are often taught to offer prognoses coldly, ensuring that patients have a clear sense of how long they have left and to help inform their treatment choices. And yet, this training does not necessarily align with what patients want and need. For instance, in a recent survey of patients with chronic obstructive pulmonary disease, patients consistently rated physicians poorly at discussing prognosis, what dying might be like, as well as spirituality and religion.

But at the same time, these matter-of-fact statements about death probably help protect us. Death is a routine, inevitable part of an oncologist’s life, and over time, oncology training and practice hardens us to it. During medical school, I remember that a patient dying would trigger immediate reflection, sadness, and conversation with our peers. Now, unless I know a patient well, I find myself rarely reflecting on the patient behind the facts. This evolution is natural for an oncologist: If you don’t develop a tough skin about death, you may become overwhelmed with the frequency of it.

The COVID pandemic has amped our hardness toward death into overdrive. Whether we are in the intensive care unit or simply viewing death rates during the most recent COVID Delta wave, many of us cope by disassociating a face from a name.
 

 

 

Making time for reflection

But taking time to reflect can be therapeutic.

I recently referred a patient with metastatic prostate cancer for a phase 1 trial at an outside institution. He was one of the first patients in my genitourinary malignancies clinic when I started as an attending. The patient had progressed through several lines of therapy and was being referred for an investigational phase 1 therapy. We had discussed hospice referral, and the patient was ready for it if this therapy didn’t work out.

I did not see or hear from the man while he was on the trial. A few months later, however, the principal investigator of the trial called me to let me know the patient had progressed through the agent, suffering from significant urinary obstruction, and he was on hospice. “Unfortunately,” the investigator told me, “he’s not going to live much longer.”

When I checked in with the hospice, the patient had died.

I was surprised again at how matter-of-fact the discussion of death had been. But I was even more surprised by my own reaction. Despite the relationship I had formed with the patient, I did not feel much when I heard he had died. I didn’t have time to process the news in the moment. It was time to move on to the next patient.

It was only later, when I called the patient’s family, that I allowed my emotions to flood in. I told his family how grateful I was to know him, how strong he’d been. The patient’s family and I talked about the human, not his passing. It felt good.
 

Abandoning locker room talk

So how do we change how we talk about death? I don’t think the answer is massive educational programs or passing responsibility for advance care planning onto palliative care specialists. The change needs to be driven by individual oncologists. We can call out discussions of death that make us uncomfortable, gently reminding each other that we’re talking about a human life.

We can learn from our palliative care colleagues; their conversations about death routinely include a patient’s support system and personal stories. Palliative care doctors always refer to the patient by name, which helps humanize the person behind the chart.

We can emphasize a feeling of hope, a sentiment that may also be therapeutic to our patients. Even when a patient is dying, there is always something to be done. We can comfort their family, explaining what brought us to this point and how sorry we are that this is happening. We can provide options for symptom control and help patients manage those symptoms.

And we can allow ourselves to talk about how much a death affects us. We can acknowledge how much it sucks that a patient is going to die, how challenging that will be to his/her family, and how we wish it could have ended differently.

Subtle changes like these will improve our own ability to process and discuss death and will ultimately lead to better relationships with our patients. But it starts with eliminating the “locker room talk” of how we discuss death.
 

Ravi B. Parikh, MD, MPP, is a medical oncologist and faculty member at the University of Pennsylvania and the Philadelphia VA Medical Center, an adjunct fellow at the Leonard Davis Institute of Health Economics, and senior clinical advisor at the Coalition to Transform Advanced Care (C-TAC). He has served as a director, officer, partner, employee, adviser, consultant, or trustee for GNS Healthcare, Nanology, and Cancer Study Group, and he has received research grant from Embedded Healthcare, Veterans Administration, PCF, National Palliative Care Research Center, and MUSC. A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Immunotherapy Experience in Nonagenarian Veterans with Cancer

Article Type
Changed
Thu, 12/15/2022 - 14:36

Background

 Immune checkpoint inhibitors [cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death 1 receptor/ programmed death ligand-1 (PD1/ PD-L1)] have received broad FDA approval in most cancers. As clinical use of these agents proliferates, data for their efficacy and safety in elderly populations, particularly nonagenarians, is sparse [1]. Nonagenarians are commonly excluded from or underrepresented in clinical trials. This occurs despite the fact that the elderly embody the fastest growing portion of the population worldwide [2]. The purpose of this project was to describe the experience of treating veterans >/= 90 years of age with immune checkpoint inhibitors (IPI) for cancer.

Methods

We reviewed drug exposure in Nonagenarians who received IPI within the VA system nationwide between 2016-2017 using CAPRI. We identified 48 veterans and reviewed each patient’s treatment, duration of immunotherapy exposure, response, and toxicity to generate a global review on how those nonagenarians tolerated treatment. Demographic data of study participants and all endpoints have been analyzed using descriptive statistics.

Results

We obtained the record data for 48 veterans who received CPI in the VA health system between 2016 and 2017. Baseline characteristics revealed that the majority of patients (N=26) were ECOG 0-1 at the time of treatment initiation. The most commonly treated malignancies included melanoma (N=19) and NSCLC (N=15) with the majority of cancers being stage IV (N=42). The primary outcome measures are duration of therapy (average 12.2 cycles) and overall survival (average 1.59 years). The secondary outcome is adverse events, with a total rate of 27.1% and grade III/IV events occurring at a rate of 6.3%

Implications

These cases and data points illustrate that immunotherapy is being used in nonagenarians. With close monitoring of toxicities, nonagenarians with acceptable performance status can be treated with immunotherapy with their consent. Future aims will focus on the addition of more data points by expanding to include 2018.

 

References

1. Lewis JH, Kilgore ML, Goldman DP, Trimble EL, Kaplan R,Montello MJ, et al. Participation of patients 65 years of age or older in cancer clinical trials. J Clin Oncol. 2003;21(7):1383–9. 2. Sgambato S, Casaluce F, Gridelli C. The role of checkpoint inhibitors immunotherapy in advanced non-small cell lung cancer in the elderly. Expert Opin Biol Ther. 2017;17(5):565-571.

Issue
Federal Practitioner - 38(4)s
Publications
Topics
Page Number
S11
Sections

Background

 Immune checkpoint inhibitors [cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death 1 receptor/ programmed death ligand-1 (PD1/ PD-L1)] have received broad FDA approval in most cancers. As clinical use of these agents proliferates, data for their efficacy and safety in elderly populations, particularly nonagenarians, is sparse [1]. Nonagenarians are commonly excluded from or underrepresented in clinical trials. This occurs despite the fact that the elderly embody the fastest growing portion of the population worldwide [2]. The purpose of this project was to describe the experience of treating veterans >/= 90 years of age with immune checkpoint inhibitors (IPI) for cancer.

Methods

We reviewed drug exposure in Nonagenarians who received IPI within the VA system nationwide between 2016-2017 using CAPRI. We identified 48 veterans and reviewed each patient’s treatment, duration of immunotherapy exposure, response, and toxicity to generate a global review on how those nonagenarians tolerated treatment. Demographic data of study participants and all endpoints have been analyzed using descriptive statistics.

Results

We obtained the record data for 48 veterans who received CPI in the VA health system between 2016 and 2017. Baseline characteristics revealed that the majority of patients (N=26) were ECOG 0-1 at the time of treatment initiation. The most commonly treated malignancies included melanoma (N=19) and NSCLC (N=15) with the majority of cancers being stage IV (N=42). The primary outcome measures are duration of therapy (average 12.2 cycles) and overall survival (average 1.59 years). The secondary outcome is adverse events, with a total rate of 27.1% and grade III/IV events occurring at a rate of 6.3%

Implications

These cases and data points illustrate that immunotherapy is being used in nonagenarians. With close monitoring of toxicities, nonagenarians with acceptable performance status can be treated with immunotherapy with their consent. Future aims will focus on the addition of more data points by expanding to include 2018.

 

References

1. Lewis JH, Kilgore ML, Goldman DP, Trimble EL, Kaplan R,Montello MJ, et al. Participation of patients 65 years of age or older in cancer clinical trials. J Clin Oncol. 2003;21(7):1383–9. 2. Sgambato S, Casaluce F, Gridelli C. The role of checkpoint inhibitors immunotherapy in advanced non-small cell lung cancer in the elderly. Expert Opin Biol Ther. 2017;17(5):565-571.

Background

 Immune checkpoint inhibitors [cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death 1 receptor/ programmed death ligand-1 (PD1/ PD-L1)] have received broad FDA approval in most cancers. As clinical use of these agents proliferates, data for their efficacy and safety in elderly populations, particularly nonagenarians, is sparse [1]. Nonagenarians are commonly excluded from or underrepresented in clinical trials. This occurs despite the fact that the elderly embody the fastest growing portion of the population worldwide [2]. The purpose of this project was to describe the experience of treating veterans >/= 90 years of age with immune checkpoint inhibitors (IPI) for cancer.

Methods

We reviewed drug exposure in Nonagenarians who received IPI within the VA system nationwide between 2016-2017 using CAPRI. We identified 48 veterans and reviewed each patient’s treatment, duration of immunotherapy exposure, response, and toxicity to generate a global review on how those nonagenarians tolerated treatment. Demographic data of study participants and all endpoints have been analyzed using descriptive statistics.

Results

We obtained the record data for 48 veterans who received CPI in the VA health system between 2016 and 2017. Baseline characteristics revealed that the majority of patients (N=26) were ECOG 0-1 at the time of treatment initiation. The most commonly treated malignancies included melanoma (N=19) and NSCLC (N=15) with the majority of cancers being stage IV (N=42). The primary outcome measures are duration of therapy (average 12.2 cycles) and overall survival (average 1.59 years). The secondary outcome is adverse events, with a total rate of 27.1% and grade III/IV events occurring at a rate of 6.3%

Implications

These cases and data points illustrate that immunotherapy is being used in nonagenarians. With close monitoring of toxicities, nonagenarians with acceptable performance status can be treated with immunotherapy with their consent. Future aims will focus on the addition of more data points by expanding to include 2018.

 

References

1. Lewis JH, Kilgore ML, Goldman DP, Trimble EL, Kaplan R,Montello MJ, et al. Participation of patients 65 years of age or older in cancer clinical trials. J Clin Oncol. 2003;21(7):1383–9. 2. Sgambato S, Casaluce F, Gridelli C. The role of checkpoint inhibitors immunotherapy in advanced non-small cell lung cancer in the elderly. Expert Opin Biol Ther. 2017;17(5):565-571.

Issue
Federal Practitioner - 38(4)s
Issue
Federal Practitioner - 38(4)s
Page Number
S11
Page Number
S11
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 09/23/2021 - 15:30
Un-Gate On Date
Thu, 09/23/2021 - 15:30
Use ProPublica
CFC Schedule Remove Status
Thu, 09/23/2021 - 15:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Evaluation of the Impact of the VHA National Precision Oncology Program (NPOP) on Prior Authorization Adjudication of Targeted Anti-Cancer Agents

Article Type
Changed
Thu, 12/15/2022 - 14:36

Purpose

To evaluate the impact of the VHA NPOP on prescribing and prior authorization approval of targeted anti-cancer therapies.

Background

Comprehensive genomic profiling (CGP) next-generation sequencing (NGS) panels have seen increased use to guide oncology therapeutic decision making. In-line with the White House Cancer Moonshot initiative, the VHA established the National Precision Oncology Program (NPOP) in July of 2016 to provide veterans with easier access to CGP and help match patients with commercially available targeted oncology therapies based on their tumor molecular profile.

Methods/Data Analysis 

A retrospective review within the VHA was conducted on patients who underwent CGP testing through the VHA NPOP from July 2016 through December 2020. Prior authorization drug request (PADR) consults for targeted oncology therapies for which CGP is a companion diagnostic for use were queried and approval outcomes were determined. NPOP interfacility consult (IFC) data was queried and matched to PADR and prescription data to determine if the IFC therapy recommendation was accepted and prescribed. Descriptive statistics were used to describe patient demographics and characterize PADR and IFC outcomes.

Results 

From July 2016 to December 2020, 16,312 tumor and blood samples from 130 unique VA medical centers representing 15,467 veterans were analyzed. Approximately 15% of veterans were prescribed targeted oncology therapies that required a PADR with a 95% approval rate. Targeted therapy recommendations with corresponding level of evidence was seen in 160 of 425 IFCs. Among 160 IFCs with targeted therapy recommendations, 75 had the recommendations accepted with two denied by PADR after local review. Recommended therapies were ultimately received by 72 patients as one patient did not have an active drug order.

 

Implications

Implementation of the VHA NPOP has increased access to CGP for more than 15,000 veterans. Availability of CGP results may have affected PADR approval outcomes of targeted therapies in approximately 15% of veterans. Approximately 50% of IFCs led to approval and subsequent prescribing of recommended therapies. Further analysis of these data and trends may help guide future prescribing practices and aid with development of clinical pathways involving molecularly targeted anti-cancer therapies.

Author and Disclosure Information

Durham Veterans Affairs Health Care System, W.G. (Bill) Hefner VA Medical Center, VA National Oncology Program, Duke University School of Medicine

Issue
Federal Practitioner - 38(4)s
Publications
Topics
Page Number
S10
Sections
Author and Disclosure Information

Durham Veterans Affairs Health Care System, W.G. (Bill) Hefner VA Medical Center, VA National Oncology Program, Duke University School of Medicine

Author and Disclosure Information

Durham Veterans Affairs Health Care System, W.G. (Bill) Hefner VA Medical Center, VA National Oncology Program, Duke University School of Medicine

Purpose

To evaluate the impact of the VHA NPOP on prescribing and prior authorization approval of targeted anti-cancer therapies.

Background

Comprehensive genomic profiling (CGP) next-generation sequencing (NGS) panels have seen increased use to guide oncology therapeutic decision making. In-line with the White House Cancer Moonshot initiative, the VHA established the National Precision Oncology Program (NPOP) in July of 2016 to provide veterans with easier access to CGP and help match patients with commercially available targeted oncology therapies based on their tumor molecular profile.

Methods/Data Analysis 

A retrospective review within the VHA was conducted on patients who underwent CGP testing through the VHA NPOP from July 2016 through December 2020. Prior authorization drug request (PADR) consults for targeted oncology therapies for which CGP is a companion diagnostic for use were queried and approval outcomes were determined. NPOP interfacility consult (IFC) data was queried and matched to PADR and prescription data to determine if the IFC therapy recommendation was accepted and prescribed. Descriptive statistics were used to describe patient demographics and characterize PADR and IFC outcomes.

Results 

From July 2016 to December 2020, 16,312 tumor and blood samples from 130 unique VA medical centers representing 15,467 veterans were analyzed. Approximately 15% of veterans were prescribed targeted oncology therapies that required a PADR with a 95% approval rate. Targeted therapy recommendations with corresponding level of evidence was seen in 160 of 425 IFCs. Among 160 IFCs with targeted therapy recommendations, 75 had the recommendations accepted with two denied by PADR after local review. Recommended therapies were ultimately received by 72 patients as one patient did not have an active drug order.

 

Implications

Implementation of the VHA NPOP has increased access to CGP for more than 15,000 veterans. Availability of CGP results may have affected PADR approval outcomes of targeted therapies in approximately 15% of veterans. Approximately 50% of IFCs led to approval and subsequent prescribing of recommended therapies. Further analysis of these data and trends may help guide future prescribing practices and aid with development of clinical pathways involving molecularly targeted anti-cancer therapies.

Purpose

To evaluate the impact of the VHA NPOP on prescribing and prior authorization approval of targeted anti-cancer therapies.

Background

Comprehensive genomic profiling (CGP) next-generation sequencing (NGS) panels have seen increased use to guide oncology therapeutic decision making. In-line with the White House Cancer Moonshot initiative, the VHA established the National Precision Oncology Program (NPOP) in July of 2016 to provide veterans with easier access to CGP and help match patients with commercially available targeted oncology therapies based on their tumor molecular profile.

Methods/Data Analysis 

A retrospective review within the VHA was conducted on patients who underwent CGP testing through the VHA NPOP from July 2016 through December 2020. Prior authorization drug request (PADR) consults for targeted oncology therapies for which CGP is a companion diagnostic for use were queried and approval outcomes were determined. NPOP interfacility consult (IFC) data was queried and matched to PADR and prescription data to determine if the IFC therapy recommendation was accepted and prescribed. Descriptive statistics were used to describe patient demographics and characterize PADR and IFC outcomes.

Results 

From July 2016 to December 2020, 16,312 tumor and blood samples from 130 unique VA medical centers representing 15,467 veterans were analyzed. Approximately 15% of veterans were prescribed targeted oncology therapies that required a PADR with a 95% approval rate. Targeted therapy recommendations with corresponding level of evidence was seen in 160 of 425 IFCs. Among 160 IFCs with targeted therapy recommendations, 75 had the recommendations accepted with two denied by PADR after local review. Recommended therapies were ultimately received by 72 patients as one patient did not have an active drug order.

 

Implications

Implementation of the VHA NPOP has increased access to CGP for more than 15,000 veterans. Availability of CGP results may have affected PADR approval outcomes of targeted therapies in approximately 15% of veterans. Approximately 50% of IFCs led to approval and subsequent prescribing of recommended therapies. Further analysis of these data and trends may help guide future prescribing practices and aid with development of clinical pathways involving molecularly targeted anti-cancer therapies.

Issue
Federal Practitioner - 38(4)s
Issue
Federal Practitioner - 38(4)s
Page Number
S10
Page Number
S10
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 09/23/2021 - 14:15
Un-Gate On Date
Thu, 09/23/2021 - 14:15
Use ProPublica
CFC Schedule Remove Status
Thu, 09/23/2021 - 14:15
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Improving the Efficiency of Ordering Next Generation Sequencing During New Patient Triage: A Quality Improvement Project

Article Type
Changed
Thu, 12/15/2022 - 14:36

Objective

To decrease the time to treatment by streamlining ordering of next generation sequencing (NGS) during new patient triage utilizing a centralized document of indications for testing.

Background

Use of NGS in management of patients with cancer is rapidly expanding. In 2017, over 75% of oncologists reported using NGS to guide treatment decisions (1). NGS testing is also now incorporated into 67% of NCCN guidelines (2). However, due to the wide variety and changing indications for NGS, integrating testing into routine clinical care can be challenging.

Results

A total of 118 new patients were seen at the SLC VA Oncology Clinic between 2020-2021 of which 21 met criteria for NGS testing at time of triage consult, 10 before and 11 after the intervention. Median time from triage to treatment initiation was 30 days (30-33) after the incorporation of the document into clinic workflow compared to 63 days (47-66). Median time from biopsy to NGS results was similar between pre- and post-intervention groups, 28 (25-49) vs 26 days (18.5-26.5).

Conclusion

Our centralized summary of NGS indications is easily updated and accessible to staff. To date, shorter times from triage to treatment have been seen after integrating this document into clinic workflow. As our sample size is small, further evaluation of this trend is required. However, our data suggests that additional improvement may be achieved through incorporating this document into the Pathology department’s workflow.

 

References

(1) Freedman A et al. Use of NGS sequencing tests to guide cancer treatment: results from a nationally representative survey of oncologists in the United States. JCO Precis Oncol. 2018;2:1-13. (2) Conway J et al. NGS and the clinical oncology workflow: data challenges, proposed solutions and a call to action. JCO Precis Oncol. 2019;3:1-10.

Issue
Federal Practitioner - 38(4)s
Publications
Topics
Page Number
S10
Sections

Objective

To decrease the time to treatment by streamlining ordering of next generation sequencing (NGS) during new patient triage utilizing a centralized document of indications for testing.

Background

Use of NGS in management of patients with cancer is rapidly expanding. In 2017, over 75% of oncologists reported using NGS to guide treatment decisions (1). NGS testing is also now incorporated into 67% of NCCN guidelines (2). However, due to the wide variety and changing indications for NGS, integrating testing into routine clinical care can be challenging.

Results

A total of 118 new patients were seen at the SLC VA Oncology Clinic between 2020-2021 of which 21 met criteria for NGS testing at time of triage consult, 10 before and 11 after the intervention. Median time from triage to treatment initiation was 30 days (30-33) after the incorporation of the document into clinic workflow compared to 63 days (47-66). Median time from biopsy to NGS results was similar between pre- and post-intervention groups, 28 (25-49) vs 26 days (18.5-26.5).

Conclusion

Our centralized summary of NGS indications is easily updated and accessible to staff. To date, shorter times from triage to treatment have been seen after integrating this document into clinic workflow. As our sample size is small, further evaluation of this trend is required. However, our data suggests that additional improvement may be achieved through incorporating this document into the Pathology department’s workflow.

 

References

(1) Freedman A et al. Use of NGS sequencing tests to guide cancer treatment: results from a nationally representative survey of oncologists in the United States. JCO Precis Oncol. 2018;2:1-13. (2) Conway J et al. NGS and the clinical oncology workflow: data challenges, proposed solutions and a call to action. JCO Precis Oncol. 2019;3:1-10.

Objective

To decrease the time to treatment by streamlining ordering of next generation sequencing (NGS) during new patient triage utilizing a centralized document of indications for testing.

Background

Use of NGS in management of patients with cancer is rapidly expanding. In 2017, over 75% of oncologists reported using NGS to guide treatment decisions (1). NGS testing is also now incorporated into 67% of NCCN guidelines (2). However, due to the wide variety and changing indications for NGS, integrating testing into routine clinical care can be challenging.

Results

A total of 118 new patients were seen at the SLC VA Oncology Clinic between 2020-2021 of which 21 met criteria for NGS testing at time of triage consult, 10 before and 11 after the intervention. Median time from triage to treatment initiation was 30 days (30-33) after the incorporation of the document into clinic workflow compared to 63 days (47-66). Median time from biopsy to NGS results was similar between pre- and post-intervention groups, 28 (25-49) vs 26 days (18.5-26.5).

Conclusion

Our centralized summary of NGS indications is easily updated and accessible to staff. To date, shorter times from triage to treatment have been seen after integrating this document into clinic workflow. As our sample size is small, further evaluation of this trend is required. However, our data suggests that additional improvement may be achieved through incorporating this document into the Pathology department’s workflow.

 

References

(1) Freedman A et al. Use of NGS sequencing tests to guide cancer treatment: results from a nationally representative survey of oncologists in the United States. JCO Precis Oncol. 2018;2:1-13. (2) Conway J et al. NGS and the clinical oncology workflow: data challenges, proposed solutions and a call to action. JCO Precis Oncol. 2019;3:1-10.

Issue
Federal Practitioner - 38(4)s
Issue
Federal Practitioner - 38(4)s
Page Number
S10
Page Number
S10
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 09/23/2021 - 14:00
Un-Gate On Date
Thu, 09/23/2021 - 14:00
Use ProPublica
CFC Schedule Remove Status
Thu, 09/23/2021 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article