Duty to Assist: Assisting Veterans With Exposures to Hazardous Materials

Article Type
Changed
Thu, 12/15/2022 - 14:27

Background

Veterans are not well informed of the presumptive conditions associated with toxic exposures endured during military service. A quality improvement project was created to increase awareness. The purpose of this project was to: Raise awareness of presumptive conditions associated with Agent Orange, Camp LeJeune contaminated water and Southwest Asia Burn Pit (fine particulate) exposure, to notify veterans how to initiate the disability benefits application process, and to inform veterans and providers of new changes in legislation (ie, Blue Water and Southwest Asia).

Methods

Using the Cancer Registry, the cancer access coordinator identified veterans with a diagnosis on each of the 3 presumptive condition lists for Agent Orange, Camp Lejeune, and Southwest Asia. These lists were then forwarded to informatics who further screened for military service history. Informative mailers were then sent to the identified veterans, alerting them to their potential eligibility for disability benefits. In addition, the mailers informed veterans how to initiate the benefits application process, how to schedule Disability Benefit Questionnaire (DBQ) exams to expedite the process, as well as contact information for local Veteran’s Service Commission (VSC) and the Veterans Benefits Administration (VBA) for further assistance. These letters were also distributed throughout the medical facility and was shared at cancer committee meetings to increase provider awareness.

Results

In 2021, 604 veterans were identified as potentially eligible for disability benefits and were contacted via mailer. As a result, 153 veterans have been granted service-connected benefits for their identified condition. An additional 91 mailers have been sent since January 2022.

Conclusions

Utilizing this simple practice increases both veteran and provider awareness of presumptive conditions, and aids in veterans receiving the disability compensation they are entitled to. In addition, this practice improves the overall quality of care the veterans receive through the VA and gives us a chance to give back to our veterans.

Publications
Topics
Sections

Background

Veterans are not well informed of the presumptive conditions associated with toxic exposures endured during military service. A quality improvement project was created to increase awareness. The purpose of this project was to: Raise awareness of presumptive conditions associated with Agent Orange, Camp LeJeune contaminated water and Southwest Asia Burn Pit (fine particulate) exposure, to notify veterans how to initiate the disability benefits application process, and to inform veterans and providers of new changes in legislation (ie, Blue Water and Southwest Asia).

Methods

Using the Cancer Registry, the cancer access coordinator identified veterans with a diagnosis on each of the 3 presumptive condition lists for Agent Orange, Camp Lejeune, and Southwest Asia. These lists were then forwarded to informatics who further screened for military service history. Informative mailers were then sent to the identified veterans, alerting them to their potential eligibility for disability benefits. In addition, the mailers informed veterans how to initiate the benefits application process, how to schedule Disability Benefit Questionnaire (DBQ) exams to expedite the process, as well as contact information for local Veteran’s Service Commission (VSC) and the Veterans Benefits Administration (VBA) for further assistance. These letters were also distributed throughout the medical facility and was shared at cancer committee meetings to increase provider awareness.

Results

In 2021, 604 veterans were identified as potentially eligible for disability benefits and were contacted via mailer. As a result, 153 veterans have been granted service-connected benefits for their identified condition. An additional 91 mailers have been sent since January 2022.

Conclusions

Utilizing this simple practice increases both veteran and provider awareness of presumptive conditions, and aids in veterans receiving the disability compensation they are entitled to. In addition, this practice improves the overall quality of care the veterans receive through the VA and gives us a chance to give back to our veterans.

Background

Veterans are not well informed of the presumptive conditions associated with toxic exposures endured during military service. A quality improvement project was created to increase awareness. The purpose of this project was to: Raise awareness of presumptive conditions associated with Agent Orange, Camp LeJeune contaminated water and Southwest Asia Burn Pit (fine particulate) exposure, to notify veterans how to initiate the disability benefits application process, and to inform veterans and providers of new changes in legislation (ie, Blue Water and Southwest Asia).

Methods

Using the Cancer Registry, the cancer access coordinator identified veterans with a diagnosis on each of the 3 presumptive condition lists for Agent Orange, Camp Lejeune, and Southwest Asia. These lists were then forwarded to informatics who further screened for military service history. Informative mailers were then sent to the identified veterans, alerting them to their potential eligibility for disability benefits. In addition, the mailers informed veterans how to initiate the benefits application process, how to schedule Disability Benefit Questionnaire (DBQ) exams to expedite the process, as well as contact information for local Veteran’s Service Commission (VSC) and the Veterans Benefits Administration (VBA) for further assistance. These letters were also distributed throughout the medical facility and was shared at cancer committee meetings to increase provider awareness.

Results

In 2021, 604 veterans were identified as potentially eligible for disability benefits and were contacted via mailer. As a result, 153 veterans have been granted service-connected benefits for their identified condition. An additional 91 mailers have been sent since January 2022.

Conclusions

Utilizing this simple practice increases both veteran and provider awareness of presumptive conditions, and aids in veterans receiving the disability compensation they are entitled to. In addition, this practice improves the overall quality of care the veterans receive through the VA and gives us a chance to give back to our veterans.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/31/2022 - 15:00
Un-Gate On Date
Wed, 08/31/2022 - 15:00
Use ProPublica
CFC Schedule Remove Status
Wed, 08/31/2022 - 15:00
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

Online tool IDs people with genetic mutations linked to cancer

Article Type
Changed
Thu, 12/15/2022 - 14:27

A new online tool can quickly and accurately identify individuals who may benefit from genetic testing because they likely carry pathogenic germline variants (PGVs) in a diverse spectrum of cancer susceptibility genes.

The PREMMplus online tool was developed and validated by researchers at the Dana-Farber Cancer Institute, Boston using three cohorts involving more than 30,000 individuals who had undergone multigene hereditary cancer risk testing.

The study was published online  in the Journal of Clinical Oncology.

“Our findings show that PREMMplus has the potential to change the model by which patients and family members are referred for genetic testing and counseling,” senior author Sapna Syngal, MD, MPH, with Dana-Farber/Brigham and Women’s Hospital, Boston, said in an institution news release.

Traditionally, when there is concern about a family cancer history, the individual is referred to a genetics clinic, where a counselor takes a complete family history.

“At a time when there’s a shortage of genetic counselors, PREMMplus can help streamline risk assessment and ensure that their time can be focused on where they’re most needed – helping people understand the results of genetic testing and the options available when a cancer-susceptibility gene is found,” Dr. Syngal says.
 

Online tool

The tool uses clinical data (age, sex, ethnicity, and personal/family history of 18 cancers) to determine an individual’s likelihood of harboring a PGV in 19 cancer susceptibility genes.

A PREMMplus score of 2.5% or greater had a 89%-94% sensitivity and > 97% negative predictive value (NPV) for identifying individuals with PGVs in 11 well-defined “category A” high-penetrance cancer risk genes: APC, BRCA1, BRCA2, CDH1, EPCAM, MLH1, MSH2, MSH6, biallelic MUTYH, PMS2, and TP53.

These PGVs “represent diverse types of inherited cancer risk for which there are established risk-reduction guidelines,” the study team says. Cancers associated with these PGVs include breast, ovarian, colorectal, pancreatic, and prostate cancer, as well as those that make up Lynch syndrome.

The ability of PREMMplus to identify individuals with PGVs in “moderate-penetrance” cancer risk genes (such as CHEK2 and ATM) was somewhat reduced but was still “quite strong” (84%-90% sensitivity and > 93% NPV), the study team reports.

In an interview, Dr. Syngal said her ultimate vision of this online tool is that it will be adapted into the electronic medical record (EMR).

“Through the EMR, it might somehow get pushed out to people before an oncology or primary care appointment or before a mammography or colonoscopy. Then by the time they come in, the doctor or nurse practitioner has the information and can refer them for genetic testing if appropriate,” Dr. Syngal explained.

The tool is not currently available for routine clinical use. The goal is to make it available online in a couple of months.

Dr. Syngal said two versions will be available. One will be a user-friendly version that can be filled out directly by patients and that will tell whether someone passes the threshold of needing genetic testing. The patient would then take that information to their primary care doctor.

With the second version, the doctor and patient would fill out the information together during an office visit.

PREMMplus would be free for the individual patient or provider.

“What we hope is that hospital systems will use it and that insurance companies will also use it as a way to say who needs testing and who to approve for testing,” Dr. Syngal told this news organization.

“For a hospital system or a genetic testing company, for example, that wants to integrate it into their direct-to-consumer platform, they would have to take out a license from Dana-Farber, and cost would be negotiated with each entity based on how they’re going to use it,” Dr. Syngal said.

Funding for the research was provided by the National Institutes of Health. A complete list of author disclosures is available with the original article.

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

Publications
Topics
Sections

A new online tool can quickly and accurately identify individuals who may benefit from genetic testing because they likely carry pathogenic germline variants (PGVs) in a diverse spectrum of cancer susceptibility genes.

The PREMMplus online tool was developed and validated by researchers at the Dana-Farber Cancer Institute, Boston using three cohorts involving more than 30,000 individuals who had undergone multigene hereditary cancer risk testing.

The study was published online  in the Journal of Clinical Oncology.

“Our findings show that PREMMplus has the potential to change the model by which patients and family members are referred for genetic testing and counseling,” senior author Sapna Syngal, MD, MPH, with Dana-Farber/Brigham and Women’s Hospital, Boston, said in an institution news release.

Traditionally, when there is concern about a family cancer history, the individual is referred to a genetics clinic, where a counselor takes a complete family history.

“At a time when there’s a shortage of genetic counselors, PREMMplus can help streamline risk assessment and ensure that their time can be focused on where they’re most needed – helping people understand the results of genetic testing and the options available when a cancer-susceptibility gene is found,” Dr. Syngal says.
 

Online tool

The tool uses clinical data (age, sex, ethnicity, and personal/family history of 18 cancers) to determine an individual’s likelihood of harboring a PGV in 19 cancer susceptibility genes.

A PREMMplus score of 2.5% or greater had a 89%-94% sensitivity and > 97% negative predictive value (NPV) for identifying individuals with PGVs in 11 well-defined “category A” high-penetrance cancer risk genes: APC, BRCA1, BRCA2, CDH1, EPCAM, MLH1, MSH2, MSH6, biallelic MUTYH, PMS2, and TP53.

These PGVs “represent diverse types of inherited cancer risk for which there are established risk-reduction guidelines,” the study team says. Cancers associated with these PGVs include breast, ovarian, colorectal, pancreatic, and prostate cancer, as well as those that make up Lynch syndrome.

The ability of PREMMplus to identify individuals with PGVs in “moderate-penetrance” cancer risk genes (such as CHEK2 and ATM) was somewhat reduced but was still “quite strong” (84%-90% sensitivity and > 93% NPV), the study team reports.

In an interview, Dr. Syngal said her ultimate vision of this online tool is that it will be adapted into the electronic medical record (EMR).

“Through the EMR, it might somehow get pushed out to people before an oncology or primary care appointment or before a mammography or colonoscopy. Then by the time they come in, the doctor or nurse practitioner has the information and can refer them for genetic testing if appropriate,” Dr. Syngal explained.

The tool is not currently available for routine clinical use. The goal is to make it available online in a couple of months.

Dr. Syngal said two versions will be available. One will be a user-friendly version that can be filled out directly by patients and that will tell whether someone passes the threshold of needing genetic testing. The patient would then take that information to their primary care doctor.

With the second version, the doctor and patient would fill out the information together during an office visit.

PREMMplus would be free for the individual patient or provider.

“What we hope is that hospital systems will use it and that insurance companies will also use it as a way to say who needs testing and who to approve for testing,” Dr. Syngal told this news organization.

“For a hospital system or a genetic testing company, for example, that wants to integrate it into their direct-to-consumer platform, they would have to take out a license from Dana-Farber, and cost would be negotiated with each entity based on how they’re going to use it,” Dr. Syngal said.

Funding for the research was provided by the National Institutes of Health. A complete list of author disclosures is available with the original article.

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

A new online tool can quickly and accurately identify individuals who may benefit from genetic testing because they likely carry pathogenic germline variants (PGVs) in a diverse spectrum of cancer susceptibility genes.

The PREMMplus online tool was developed and validated by researchers at the Dana-Farber Cancer Institute, Boston using three cohorts involving more than 30,000 individuals who had undergone multigene hereditary cancer risk testing.

The study was published online  in the Journal of Clinical Oncology.

“Our findings show that PREMMplus has the potential to change the model by which patients and family members are referred for genetic testing and counseling,” senior author Sapna Syngal, MD, MPH, with Dana-Farber/Brigham and Women’s Hospital, Boston, said in an institution news release.

Traditionally, when there is concern about a family cancer history, the individual is referred to a genetics clinic, where a counselor takes a complete family history.

“At a time when there’s a shortage of genetic counselors, PREMMplus can help streamline risk assessment and ensure that their time can be focused on where they’re most needed – helping people understand the results of genetic testing and the options available when a cancer-susceptibility gene is found,” Dr. Syngal says.
 

Online tool

The tool uses clinical data (age, sex, ethnicity, and personal/family history of 18 cancers) to determine an individual’s likelihood of harboring a PGV in 19 cancer susceptibility genes.

A PREMMplus score of 2.5% or greater had a 89%-94% sensitivity and > 97% negative predictive value (NPV) for identifying individuals with PGVs in 11 well-defined “category A” high-penetrance cancer risk genes: APC, BRCA1, BRCA2, CDH1, EPCAM, MLH1, MSH2, MSH6, biallelic MUTYH, PMS2, and TP53.

These PGVs “represent diverse types of inherited cancer risk for which there are established risk-reduction guidelines,” the study team says. Cancers associated with these PGVs include breast, ovarian, colorectal, pancreatic, and prostate cancer, as well as those that make up Lynch syndrome.

The ability of PREMMplus to identify individuals with PGVs in “moderate-penetrance” cancer risk genes (such as CHEK2 and ATM) was somewhat reduced but was still “quite strong” (84%-90% sensitivity and > 93% NPV), the study team reports.

In an interview, Dr. Syngal said her ultimate vision of this online tool is that it will be adapted into the electronic medical record (EMR).

“Through the EMR, it might somehow get pushed out to people before an oncology or primary care appointment or before a mammography or colonoscopy. Then by the time they come in, the doctor or nurse practitioner has the information and can refer them for genetic testing if appropriate,” Dr. Syngal explained.

The tool is not currently available for routine clinical use. The goal is to make it available online in a couple of months.

Dr. Syngal said two versions will be available. One will be a user-friendly version that can be filled out directly by patients and that will tell whether someone passes the threshold of needing genetic testing. The patient would then take that information to their primary care doctor.

With the second version, the doctor and patient would fill out the information together during an office visit.

PREMMplus would be free for the individual patient or provider.

“What we hope is that hospital systems will use it and that insurance companies will also use it as a way to say who needs testing and who to approve for testing,” Dr. Syngal told this news organization.

“For a hospital system or a genetic testing company, for example, that wants to integrate it into their direct-to-consumer platform, they would have to take out a license from Dana-Farber, and cost would be negotiated with each entity based on how they’re going to use it,” Dr. Syngal said.

Funding for the research was provided by the National Institutes of Health. A complete list of author disclosures is available with the original article.

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

Intera Oncology recalls hepatic artery infusion pumps for possible life-threatening issue

Article Type
Changed
Thu, 12/15/2022 - 14:27

Intera Oncology has recalled 440 Intera 3000 Hepatic Artery Infusion Pumps following three reports of potentially life-threatening medication flow rates.

Although no injuries or deaths related to the pump malfunction have been reported yet, the U.S. Food and Drug Administration has deemed the recall Class I, the most serious category that indicates the device could cause injury or death.

Intera Oncology initiated the recall in July following reports from clinicians that the pumps, which are implanted to deliver chemotherapy to treat liver tumors, were delivering medications faster than expected. A fast flow rate can lead to life-threatening hematologic toxicity, neurotoxicity, or death. It also means patients will run out of medication too soon, potentially leading to disease progression or death.

The FDA notice states the company has advised customers to continue to monitor flow rate as per standard refill procedure as well as monitor for liver toxicity to adjust dosing as per standard protocols.

The company also said to consider pump replacement if altered flow can’t be adequately managed by dosing adjustments or having patients come in for medication refills and to verify the flow rate sooner than every 2 weeks if the pump appears to be flowing more than 15% outside its labeled specification.

Questions about the recall can be directed to Intera Oncology at (800) 660-2660 or support@interaoncol.

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

Publications
Topics
Sections

Intera Oncology has recalled 440 Intera 3000 Hepatic Artery Infusion Pumps following three reports of potentially life-threatening medication flow rates.

Although no injuries or deaths related to the pump malfunction have been reported yet, the U.S. Food and Drug Administration has deemed the recall Class I, the most serious category that indicates the device could cause injury or death.

Intera Oncology initiated the recall in July following reports from clinicians that the pumps, which are implanted to deliver chemotherapy to treat liver tumors, were delivering medications faster than expected. A fast flow rate can lead to life-threatening hematologic toxicity, neurotoxicity, or death. It also means patients will run out of medication too soon, potentially leading to disease progression or death.

The FDA notice states the company has advised customers to continue to monitor flow rate as per standard refill procedure as well as monitor for liver toxicity to adjust dosing as per standard protocols.

The company also said to consider pump replacement if altered flow can’t be adequately managed by dosing adjustments or having patients come in for medication refills and to verify the flow rate sooner than every 2 weeks if the pump appears to be flowing more than 15% outside its labeled specification.

Questions about the recall can be directed to Intera Oncology at (800) 660-2660 or support@interaoncol.

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

Intera Oncology has recalled 440 Intera 3000 Hepatic Artery Infusion Pumps following three reports of potentially life-threatening medication flow rates.

Although no injuries or deaths related to the pump malfunction have been reported yet, the U.S. Food and Drug Administration has deemed the recall Class I, the most serious category that indicates the device could cause injury or death.

Intera Oncology initiated the recall in July following reports from clinicians that the pumps, which are implanted to deliver chemotherapy to treat liver tumors, were delivering medications faster than expected. A fast flow rate can lead to life-threatening hematologic toxicity, neurotoxicity, or death. It also means patients will run out of medication too soon, potentially leading to disease progression or death.

The FDA notice states the company has advised customers to continue to monitor flow rate as per standard refill procedure as well as monitor for liver toxicity to adjust dosing as per standard protocols.

The company also said to consider pump replacement if altered flow can’t be adequately managed by dosing adjustments or having patients come in for medication refills and to verify the flow rate sooner than every 2 weeks if the pump appears to be flowing more than 15% outside its labeled specification.

Questions about the recall can be directed to Intera Oncology at (800) 660-2660 or support@interaoncol.

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

Evolocumab benefits accrue with longer follow-up: FOURIER OLE

Article Type
Changed
Thu, 12/15/2022 - 14:27

Long-term lipid lowering with evolocumab (Repatha) further reduces cardiovascular events, including CV death, without a safety signal, according to results from the FOURIER open-label extension (OLE) study.

In the parent FOURIER trial, treatment with the proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor over a median of 2.2 years reduced the primary efficacy endpoint by 15% but showed no CV mortality signal, compared with placebo, in patients with atherosclerotic disease on background statin therapy.

Now with follow-up out to 8.4 years – the longest to date in any PCSK9 study – cardiovascular mortality was cut by 23% in patients who remained on evolocumab, compared with those originally assigned to placebo (3.32% vs. 4.45%; hazard ratio, 0.77; 95% confidence interval, 0.60-0.99).

The Kaplan-Meier curves during FOURIER were “essentially superimposed and it was not until the open-label extension period had begun with longer-term follow up that the benefit in terms of cardiovascular mortality reduction became apparent,” said principal investigator Michelle O’Donoghue, MD, MPH, of Brigham and Women’s Hospital, Boston.

The results were reported at the annual congress of the European Society of Cardiology and published simultaneously in Circulation.

Pivotal statin trials have median follow-up times of 4-5 years and demonstrated both a lag effect, meaning clinical benefit grew over time, and a legacy effect, where clinical benefit persisted in extended follow-up after the parent study, Dr. O’Donoghue observed.

With shorter follow-up in the parent FOURIER trial, there was evidence of a lag effect with the risk reduction in CV death, MI, and stroke increasing from 16% in the first year to 25% over time with evolocumab.

FOURIER-OLE enrolled 6,635 patients (3355 randomly assigned to evolocumab and 3280 to placebo), who completed the parent study and self-injected evolocumab subcutaneously with the choice of 140 mg every 2 weeks or 420 mg monthly. Study visits were at week 12 and then every 24 weeks. Median follow-up was 5 years.

Their mean age was 62 years, three-fourths were men, a third had diabetes. Three-fourths were on a high-intensity statin at the time of enrollment in FOURIER, and median LDL cholesterol at randomization was 91 mg/dL (2.4 mmol/L).

At week 12, the median LDL cholesterol was 30 mg/dL (0.78 mmol/L), and this was sustained throughout follow-up, Dr. O’Donoghue reported. Most patients achieved very low LDL cholesterol levels, with 63.2% achieving levels less than 40 mg/dL (1.04 mmol/L) and 26.6% less than 20 mg/dL (0.52 mmol/L).

Patients randomly assigned in the parent trial to evolocumab versus placebo had a 15% lower risk of the primary outcome of CV death, MI, stroke, hospitalization for unstable angina, or coronary revascularization (15.4% vs. 17.5%; HR, 0.85; 95% CI, 0.75-0.96).

Their risk of CV death, MI, or stroke was 20% lower (9.7% vs. 11.9%; HR, 0.80; 95% CI, 0.68-0.93), and, as noted previously, 23% lower for CV death.

When major adverse cardiovascular events data were parsed out by year, the largest LDL cholesterol reduction was in years 1 and 2 of the parent study (delta, 62 mg/dL between treatment arms), “highlighting that lag of benefit that continued to accrue with time,” Dr. O’Donoghue said.

“There was then carryover into the extension period, such that there was legacy effect from the LDL [cholesterol] delta that was seen during the parent study,” she said. “This benefit was most apparent early on during open-label extension and then, as one might expect when all patients were being treated with the same therapy, it began to attenuate somewhat with time.”

Although early studies raised concerns that very low LDL cholesterol may be associated with an increased risk of hemorrhagic stroke and neurocognitive effects, the frequency of adverse events did not increase over time with evolocumab exposure.

Annualized incidence rates for patients initially randomized to evolocumab did not exceed those for placebo-treated patients for any of the following events of interest: serious safety events (10% vs. 13%), hemorrhagic stroke (0.04% vs. 0.05%), new-onset diabetes (1.2% vs. 2.3%), muscle-related events (1.2% vs. 1.9%), injection-site reactions (0.4% vs. 0.7%), and drug-related allergic reactions (0.6% vs. 1.1%).

“Long-term use of evolocumab with a median follow-up of more than 7 years appears both safe and well tolerated,” Dr. O’Donoghue said.

Taken together with the continued accrual of cardiovascular benefit, including CV mortality, “these findings argue for early initiation of a marked and sustained LDL cholesterol reduction to maximize benefit,” she concluded.
 

 

 

Translating the benefits

Ulrich Laufs, MD, Leipzig (Germany) University Hospital, Germany, and invited commentator for the session, said the trial addresses two key issues: the long-term safety of low LDL cholesterol lowering and the long-term safety of inhibiting PCSK9, which is highly expressed not only in the liver but also in the brain, small intestine, and kidneys. Indeed, an LDL cholesterol level below 30 mg/dL is lower than the ESC treatment recommendation for very-high-risk patients and is, in fact, lower than most assays are reliable to interpret.

“So it is very important that we have these very clear data showing us that there were no adverse events, also including cataracts and hemorrhagic stroke, and these were on the level of placebo and did not increase over time,” he said.

The question of efficacy is triggered by observations of another PCSK9, the humanized monoclonal antibody bococizumab, which was associated in the SPIRE trial with an increase in LDL cholesterol over time because of neutralizing antibodies. Reassuringly, there was “completely sustained LDL [cholesterol] reduction” with no neutralizing antibodies with the fully human antibody evolocumab in FOURIER-OLE and in recent data from the OSLER-1 study, Dr. Laufs observed.

Acknowledging the potential for selection bias with an OLE program, Dr. Laufs said there are two important open questions: “Can the safety data observed for extracellular PCSK9 inhibition using an antibody be transferred to other mechanisms of PCSK9 inhibition? And obviously, from the perspective of patient care, how can we implement these important data into patient care and improve access to PCSK9 inhibitors?”

With regard to the latter point, he said physicians should be cautious in using the term “plaque regression,” opting instead for prevention and stabilization of atherosclerosis, and when using the term “legacy,” which may be misinterpreted by patients to imply there was cessation of therapy.

“From my perspective, [what] the open-label extension really shows is that earlier treatment is better,” Dr. Laufs said. “This should be our message.”

In a press conference prior to the presentation, ESC commentator Johann Bauersachs, MD, Hannover (Germany) Medical School, said “this is extremely important data because it confirms that it’s safe, and the criticism of the FOURIER study that mortality, cardiovascular mortality, was not reduced is now also reduced.”

Dr. Bauersachs said it would have been unethical to wait 7 years for a placebo-controlled trial and questioned whether data are available and suggestive of a legacy effect among patients who did not participate in the open-label extension.

Dr. O’Donoghue said unfortunately those data aren’t available but that Kaplan-Meier curves for the primary endpoint in the parent trial continued to diverge over time and that there was somewhat of a lag in terms of that divergence. “So, a median follow-up of 2 years may have been insufficient, especially for the emerging cardiovascular mortality that took longer to appear.”

The study was funded by Amgen. Dr. O’Donoghue reported receiving research grants from Amgen, AstraZeneca, Janssen, Intarcia, and Novartis, and consulting fees from Amgen, Novartis, AstraZeneca, and Janssen. Dr. Laufs reported receiving honoraria/reimbursement for lecture, study participation, and scientific cooperation with Saarland or Leipzig University, as well as relationships with multiple pharmaceutical and device makers.

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

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

Long-term lipid lowering with evolocumab (Repatha) further reduces cardiovascular events, including CV death, without a safety signal, according to results from the FOURIER open-label extension (OLE) study.

In the parent FOURIER trial, treatment with the proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor over a median of 2.2 years reduced the primary efficacy endpoint by 15% but showed no CV mortality signal, compared with placebo, in patients with atherosclerotic disease on background statin therapy.

Now with follow-up out to 8.4 years – the longest to date in any PCSK9 study – cardiovascular mortality was cut by 23% in patients who remained on evolocumab, compared with those originally assigned to placebo (3.32% vs. 4.45%; hazard ratio, 0.77; 95% confidence interval, 0.60-0.99).

The Kaplan-Meier curves during FOURIER were “essentially superimposed and it was not until the open-label extension period had begun with longer-term follow up that the benefit in terms of cardiovascular mortality reduction became apparent,” said principal investigator Michelle O’Donoghue, MD, MPH, of Brigham and Women’s Hospital, Boston.

The results were reported at the annual congress of the European Society of Cardiology and published simultaneously in Circulation.

Pivotal statin trials have median follow-up times of 4-5 years and demonstrated both a lag effect, meaning clinical benefit grew over time, and a legacy effect, where clinical benefit persisted in extended follow-up after the parent study, Dr. O’Donoghue observed.

With shorter follow-up in the parent FOURIER trial, there was evidence of a lag effect with the risk reduction in CV death, MI, and stroke increasing from 16% in the first year to 25% over time with evolocumab.

FOURIER-OLE enrolled 6,635 patients (3355 randomly assigned to evolocumab and 3280 to placebo), who completed the parent study and self-injected evolocumab subcutaneously with the choice of 140 mg every 2 weeks or 420 mg monthly. Study visits were at week 12 and then every 24 weeks. Median follow-up was 5 years.

Their mean age was 62 years, three-fourths were men, a third had diabetes. Three-fourths were on a high-intensity statin at the time of enrollment in FOURIER, and median LDL cholesterol at randomization was 91 mg/dL (2.4 mmol/L).

At week 12, the median LDL cholesterol was 30 mg/dL (0.78 mmol/L), and this was sustained throughout follow-up, Dr. O’Donoghue reported. Most patients achieved very low LDL cholesterol levels, with 63.2% achieving levels less than 40 mg/dL (1.04 mmol/L) and 26.6% less than 20 mg/dL (0.52 mmol/L).

Patients randomly assigned in the parent trial to evolocumab versus placebo had a 15% lower risk of the primary outcome of CV death, MI, stroke, hospitalization for unstable angina, or coronary revascularization (15.4% vs. 17.5%; HR, 0.85; 95% CI, 0.75-0.96).

Their risk of CV death, MI, or stroke was 20% lower (9.7% vs. 11.9%; HR, 0.80; 95% CI, 0.68-0.93), and, as noted previously, 23% lower for CV death.

When major adverse cardiovascular events data were parsed out by year, the largest LDL cholesterol reduction was in years 1 and 2 of the parent study (delta, 62 mg/dL between treatment arms), “highlighting that lag of benefit that continued to accrue with time,” Dr. O’Donoghue said.

“There was then carryover into the extension period, such that there was legacy effect from the LDL [cholesterol] delta that was seen during the parent study,” she said. “This benefit was most apparent early on during open-label extension and then, as one might expect when all patients were being treated with the same therapy, it began to attenuate somewhat with time.”

Although early studies raised concerns that very low LDL cholesterol may be associated with an increased risk of hemorrhagic stroke and neurocognitive effects, the frequency of adverse events did not increase over time with evolocumab exposure.

Annualized incidence rates for patients initially randomized to evolocumab did not exceed those for placebo-treated patients for any of the following events of interest: serious safety events (10% vs. 13%), hemorrhagic stroke (0.04% vs. 0.05%), new-onset diabetes (1.2% vs. 2.3%), muscle-related events (1.2% vs. 1.9%), injection-site reactions (0.4% vs. 0.7%), and drug-related allergic reactions (0.6% vs. 1.1%).

“Long-term use of evolocumab with a median follow-up of more than 7 years appears both safe and well tolerated,” Dr. O’Donoghue said.

Taken together with the continued accrual of cardiovascular benefit, including CV mortality, “these findings argue for early initiation of a marked and sustained LDL cholesterol reduction to maximize benefit,” she concluded.
 

 

 

Translating the benefits

Ulrich Laufs, MD, Leipzig (Germany) University Hospital, Germany, and invited commentator for the session, said the trial addresses two key issues: the long-term safety of low LDL cholesterol lowering and the long-term safety of inhibiting PCSK9, which is highly expressed not only in the liver but also in the brain, small intestine, and kidneys. Indeed, an LDL cholesterol level below 30 mg/dL is lower than the ESC treatment recommendation for very-high-risk patients and is, in fact, lower than most assays are reliable to interpret.

“So it is very important that we have these very clear data showing us that there were no adverse events, also including cataracts and hemorrhagic stroke, and these were on the level of placebo and did not increase over time,” he said.

The question of efficacy is triggered by observations of another PCSK9, the humanized monoclonal antibody bococizumab, which was associated in the SPIRE trial with an increase in LDL cholesterol over time because of neutralizing antibodies. Reassuringly, there was “completely sustained LDL [cholesterol] reduction” with no neutralizing antibodies with the fully human antibody evolocumab in FOURIER-OLE and in recent data from the OSLER-1 study, Dr. Laufs observed.

Acknowledging the potential for selection bias with an OLE program, Dr. Laufs said there are two important open questions: “Can the safety data observed for extracellular PCSK9 inhibition using an antibody be transferred to other mechanisms of PCSK9 inhibition? And obviously, from the perspective of patient care, how can we implement these important data into patient care and improve access to PCSK9 inhibitors?”

With regard to the latter point, he said physicians should be cautious in using the term “plaque regression,” opting instead for prevention and stabilization of atherosclerosis, and when using the term “legacy,” which may be misinterpreted by patients to imply there was cessation of therapy.

“From my perspective, [what] the open-label extension really shows is that earlier treatment is better,” Dr. Laufs said. “This should be our message.”

In a press conference prior to the presentation, ESC commentator Johann Bauersachs, MD, Hannover (Germany) Medical School, said “this is extremely important data because it confirms that it’s safe, and the criticism of the FOURIER study that mortality, cardiovascular mortality, was not reduced is now also reduced.”

Dr. Bauersachs said it would have been unethical to wait 7 years for a placebo-controlled trial and questioned whether data are available and suggestive of a legacy effect among patients who did not participate in the open-label extension.

Dr. O’Donoghue said unfortunately those data aren’t available but that Kaplan-Meier curves for the primary endpoint in the parent trial continued to diverge over time and that there was somewhat of a lag in terms of that divergence. “So, a median follow-up of 2 years may have been insufficient, especially for the emerging cardiovascular mortality that took longer to appear.”

The study was funded by Amgen. Dr. O’Donoghue reported receiving research grants from Amgen, AstraZeneca, Janssen, Intarcia, and Novartis, and consulting fees from Amgen, Novartis, AstraZeneca, and Janssen. Dr. Laufs reported receiving honoraria/reimbursement for lecture, study participation, and scientific cooperation with Saarland or Leipzig University, as well as relationships with multiple pharmaceutical and device makers.

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

Long-term lipid lowering with evolocumab (Repatha) further reduces cardiovascular events, including CV death, without a safety signal, according to results from the FOURIER open-label extension (OLE) study.

In the parent FOURIER trial, treatment with the proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor over a median of 2.2 years reduced the primary efficacy endpoint by 15% but showed no CV mortality signal, compared with placebo, in patients with atherosclerotic disease on background statin therapy.

Now with follow-up out to 8.4 years – the longest to date in any PCSK9 study – cardiovascular mortality was cut by 23% in patients who remained on evolocumab, compared with those originally assigned to placebo (3.32% vs. 4.45%; hazard ratio, 0.77; 95% confidence interval, 0.60-0.99).

The Kaplan-Meier curves during FOURIER were “essentially superimposed and it was not until the open-label extension period had begun with longer-term follow up that the benefit in terms of cardiovascular mortality reduction became apparent,” said principal investigator Michelle O’Donoghue, MD, MPH, of Brigham and Women’s Hospital, Boston.

The results were reported at the annual congress of the European Society of Cardiology and published simultaneously in Circulation.

Pivotal statin trials have median follow-up times of 4-5 years and demonstrated both a lag effect, meaning clinical benefit grew over time, and a legacy effect, where clinical benefit persisted in extended follow-up after the parent study, Dr. O’Donoghue observed.

With shorter follow-up in the parent FOURIER trial, there was evidence of a lag effect with the risk reduction in CV death, MI, and stroke increasing from 16% in the first year to 25% over time with evolocumab.

FOURIER-OLE enrolled 6,635 patients (3355 randomly assigned to evolocumab and 3280 to placebo), who completed the parent study and self-injected evolocumab subcutaneously with the choice of 140 mg every 2 weeks or 420 mg monthly. Study visits were at week 12 and then every 24 weeks. Median follow-up was 5 years.

Their mean age was 62 years, three-fourths were men, a third had diabetes. Three-fourths were on a high-intensity statin at the time of enrollment in FOURIER, and median LDL cholesterol at randomization was 91 mg/dL (2.4 mmol/L).

At week 12, the median LDL cholesterol was 30 mg/dL (0.78 mmol/L), and this was sustained throughout follow-up, Dr. O’Donoghue reported. Most patients achieved very low LDL cholesterol levels, with 63.2% achieving levels less than 40 mg/dL (1.04 mmol/L) and 26.6% less than 20 mg/dL (0.52 mmol/L).

Patients randomly assigned in the parent trial to evolocumab versus placebo had a 15% lower risk of the primary outcome of CV death, MI, stroke, hospitalization for unstable angina, or coronary revascularization (15.4% vs. 17.5%; HR, 0.85; 95% CI, 0.75-0.96).

Their risk of CV death, MI, or stroke was 20% lower (9.7% vs. 11.9%; HR, 0.80; 95% CI, 0.68-0.93), and, as noted previously, 23% lower for CV death.

When major adverse cardiovascular events data were parsed out by year, the largest LDL cholesterol reduction was in years 1 and 2 of the parent study (delta, 62 mg/dL between treatment arms), “highlighting that lag of benefit that continued to accrue with time,” Dr. O’Donoghue said.

“There was then carryover into the extension period, such that there was legacy effect from the LDL [cholesterol] delta that was seen during the parent study,” she said. “This benefit was most apparent early on during open-label extension and then, as one might expect when all patients were being treated with the same therapy, it began to attenuate somewhat with time.”

Although early studies raised concerns that very low LDL cholesterol may be associated with an increased risk of hemorrhagic stroke and neurocognitive effects, the frequency of adverse events did not increase over time with evolocumab exposure.

Annualized incidence rates for patients initially randomized to evolocumab did not exceed those for placebo-treated patients for any of the following events of interest: serious safety events (10% vs. 13%), hemorrhagic stroke (0.04% vs. 0.05%), new-onset diabetes (1.2% vs. 2.3%), muscle-related events (1.2% vs. 1.9%), injection-site reactions (0.4% vs. 0.7%), and drug-related allergic reactions (0.6% vs. 1.1%).

“Long-term use of evolocumab with a median follow-up of more than 7 years appears both safe and well tolerated,” Dr. O’Donoghue said.

Taken together with the continued accrual of cardiovascular benefit, including CV mortality, “these findings argue for early initiation of a marked and sustained LDL cholesterol reduction to maximize benefit,” she concluded.
 

 

 

Translating the benefits

Ulrich Laufs, MD, Leipzig (Germany) University Hospital, Germany, and invited commentator for the session, said the trial addresses two key issues: the long-term safety of low LDL cholesterol lowering and the long-term safety of inhibiting PCSK9, which is highly expressed not only in the liver but also in the brain, small intestine, and kidneys. Indeed, an LDL cholesterol level below 30 mg/dL is lower than the ESC treatment recommendation for very-high-risk patients and is, in fact, lower than most assays are reliable to interpret.

“So it is very important that we have these very clear data showing us that there were no adverse events, also including cataracts and hemorrhagic stroke, and these were on the level of placebo and did not increase over time,” he said.

The question of efficacy is triggered by observations of another PCSK9, the humanized monoclonal antibody bococizumab, which was associated in the SPIRE trial with an increase in LDL cholesterol over time because of neutralizing antibodies. Reassuringly, there was “completely sustained LDL [cholesterol] reduction” with no neutralizing antibodies with the fully human antibody evolocumab in FOURIER-OLE and in recent data from the OSLER-1 study, Dr. Laufs observed.

Acknowledging the potential for selection bias with an OLE program, Dr. Laufs said there are two important open questions: “Can the safety data observed for extracellular PCSK9 inhibition using an antibody be transferred to other mechanisms of PCSK9 inhibition? And obviously, from the perspective of patient care, how can we implement these important data into patient care and improve access to PCSK9 inhibitors?”

With regard to the latter point, he said physicians should be cautious in using the term “plaque regression,” opting instead for prevention and stabilization of atherosclerosis, and when using the term “legacy,” which may be misinterpreted by patients to imply there was cessation of therapy.

“From my perspective, [what] the open-label extension really shows is that earlier treatment is better,” Dr. Laufs said. “This should be our message.”

In a press conference prior to the presentation, ESC commentator Johann Bauersachs, MD, Hannover (Germany) Medical School, said “this is extremely important data because it confirms that it’s safe, and the criticism of the FOURIER study that mortality, cardiovascular mortality, was not reduced is now also reduced.”

Dr. Bauersachs said it would have been unethical to wait 7 years for a placebo-controlled trial and questioned whether data are available and suggestive of a legacy effect among patients who did not participate in the open-label extension.

Dr. O’Donoghue said unfortunately those data aren’t available but that Kaplan-Meier curves for the primary endpoint in the parent trial continued to diverge over time and that there was somewhat of a lag in terms of that divergence. “So, a median follow-up of 2 years may have been insufficient, especially for the emerging cardiovascular mortality that took longer to appear.”

The study was funded by Amgen. Dr. O’Donoghue reported receiving research grants from Amgen, AstraZeneca, Janssen, Intarcia, and Novartis, and consulting fees from Amgen, Novartis, AstraZeneca, and Janssen. Dr. Laufs reported receiving honoraria/reimbursement for lecture, study participation, and scientific cooperation with Saarland or Leipzig University, as well as relationships with multiple pharmaceutical and device makers.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM ESC CONGRESS 2022

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

Ketogenic Diet and Cancer: A Case Report and Feasibility Study at VA Central California Healthcare System

Article Type
Changed
Thu, 03/02/2023 - 16:53

Background

Ketogenic diet (KD) is a high-fat and low carbohydrate diet that has been reported as a treatment option for patients with cancer. KD creates a metabolic state in which blood glucose levels are reduced and ketone bodies are elevated. Cancer cells are unable to use ketone bodies for energy and metabolism due to mitochondrial dysfunction. We published the efficacy of KD in patients with cancer after failure of chemotherapy. 1 This case report is presented to evaluate the feasibility of KD concurrent with chemoimmunotherapy.

Case Report

Patient is a 69-year-old male who presented with iron deficiency anemia in 2018. Colonoscopy and biopsy showed colon adenocarcinoma. He underwent resection which confirmed stage IIIC disease. He received adjuvant treatment with FOLFOX but quickly developed pancreatic and omental metastasis. He was started on FOLFIRI + bevacizumab followed by pancreatic mass resection in 2019. Molecular testing revealed wild type KRAS, positive BRAF V600E, and high MSI. He received encorafenib + cetuximab until disease progression. Treatment was changed to pembrolizumab until PET scan showed progression. His CEA increased to 1031 in January 2021. He was subsequently started on KD concurrent with trifluridine + tipiracil + bevacizumab. He progressed after 10 months. Therapy was changed to ipilimumab + nivolumab with continuation of KD. He was strictly adherent to KD with low Glucose Ketone Index of 8.2 (confirming ketosis) but in 2022 his GKI level started to rise. His CEA, however, significantly decreased to 20 in March 2022 and PET scan showed stable disease. He presently is on maintenance nivolumab + KD while maintaining an excellent quality of life by EORTC QLQ scores.

Conclusions

The use of KD concurrently with chemotherapy and immunotherapy is still under investigation. Our case report shows that KD is tolerable with treatment and can possibly contribute to controlling progression of metastatic cancer. We are starting an investigator initiative KD trial that received a grant from R&D at VACCHCS. We will present the study protocol in poster presentation.

References

1. Tan-Shalaby JL, Carrick J, Edinger K, et al. Modified Atkins diet in advanced malignancies - final results of a safety and feasibility trial within the Veterans Affairs Pittsburgh Healthcare System]. Nutr Metab (Lond). 2016;13:52. Published 2016 Aug 12. doi:10.1186/s12986-016-0113-y

Issue
Federal Practitioner - 39(4)s
Publications
Topics
Page Number
S18
Sections

Background

Ketogenic diet (KD) is a high-fat and low carbohydrate diet that has been reported as a treatment option for patients with cancer. KD creates a metabolic state in which blood glucose levels are reduced and ketone bodies are elevated. Cancer cells are unable to use ketone bodies for energy and metabolism due to mitochondrial dysfunction. We published the efficacy of KD in patients with cancer after failure of chemotherapy. 1 This case report is presented to evaluate the feasibility of KD concurrent with chemoimmunotherapy.

Case Report

Patient is a 69-year-old male who presented with iron deficiency anemia in 2018. Colonoscopy and biopsy showed colon adenocarcinoma. He underwent resection which confirmed stage IIIC disease. He received adjuvant treatment with FOLFOX but quickly developed pancreatic and omental metastasis. He was started on FOLFIRI + bevacizumab followed by pancreatic mass resection in 2019. Molecular testing revealed wild type KRAS, positive BRAF V600E, and high MSI. He received encorafenib + cetuximab until disease progression. Treatment was changed to pembrolizumab until PET scan showed progression. His CEA increased to 1031 in January 2021. He was subsequently started on KD concurrent with trifluridine + tipiracil + bevacizumab. He progressed after 10 months. Therapy was changed to ipilimumab + nivolumab with continuation of KD. He was strictly adherent to KD with low Glucose Ketone Index of 8.2 (confirming ketosis) but in 2022 his GKI level started to rise. His CEA, however, significantly decreased to 20 in March 2022 and PET scan showed stable disease. He presently is on maintenance nivolumab + KD while maintaining an excellent quality of life by EORTC QLQ scores.

Conclusions

The use of KD concurrently with chemotherapy and immunotherapy is still under investigation. Our case report shows that KD is tolerable with treatment and can possibly contribute to controlling progression of metastatic cancer. We are starting an investigator initiative KD trial that received a grant from R&D at VACCHCS. We will present the study protocol in poster presentation.

Background

Ketogenic diet (KD) is a high-fat and low carbohydrate diet that has been reported as a treatment option for patients with cancer. KD creates a metabolic state in which blood glucose levels are reduced and ketone bodies are elevated. Cancer cells are unable to use ketone bodies for energy and metabolism due to mitochondrial dysfunction. We published the efficacy of KD in patients with cancer after failure of chemotherapy. 1 This case report is presented to evaluate the feasibility of KD concurrent with chemoimmunotherapy.

Case Report

Patient is a 69-year-old male who presented with iron deficiency anemia in 2018. Colonoscopy and biopsy showed colon adenocarcinoma. He underwent resection which confirmed stage IIIC disease. He received adjuvant treatment with FOLFOX but quickly developed pancreatic and omental metastasis. He was started on FOLFIRI + bevacizumab followed by pancreatic mass resection in 2019. Molecular testing revealed wild type KRAS, positive BRAF V600E, and high MSI. He received encorafenib + cetuximab until disease progression. Treatment was changed to pembrolizumab until PET scan showed progression. His CEA increased to 1031 in January 2021. He was subsequently started on KD concurrent with trifluridine + tipiracil + bevacizumab. He progressed after 10 months. Therapy was changed to ipilimumab + nivolumab with continuation of KD. He was strictly adherent to KD with low Glucose Ketone Index of 8.2 (confirming ketosis) but in 2022 his GKI level started to rise. His CEA, however, significantly decreased to 20 in March 2022 and PET scan showed stable disease. He presently is on maintenance nivolumab + KD while maintaining an excellent quality of life by EORTC QLQ scores.

Conclusions

The use of KD concurrently with chemotherapy and immunotherapy is still under investigation. Our case report shows that KD is tolerable with treatment and can possibly contribute to controlling progression of metastatic cancer. We are starting an investigator initiative KD trial that received a grant from R&D at VACCHCS. We will present the study protocol in poster presentation.

References

1. Tan-Shalaby JL, Carrick J, Edinger K, et al. Modified Atkins diet in advanced malignancies - final results of a safety and feasibility trial within the Veterans Affairs Pittsburgh Healthcare System]. Nutr Metab (Lond). 2016;13:52. Published 2016 Aug 12. doi:10.1186/s12986-016-0113-y

References

1. Tan-Shalaby JL, Carrick J, Edinger K, et al. Modified Atkins diet in advanced malignancies - final results of a safety and feasibility trial within the Veterans Affairs Pittsburgh Healthcare System]. Nutr Metab (Lond). 2016;13:52. Published 2016 Aug 12. doi:10.1186/s12986-016-0113-y

Issue
Federal Practitioner - 39(4)s
Issue
Federal Practitioner - 39(4)s
Page Number
S18
Page Number
S18
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 08/30/2022 - 15:15
Un-Gate On Date
Tue, 08/30/2022 - 15:15
Use ProPublica
CFC Schedule Remove Status
Tue, 08/30/2022 - 15:15
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

Capturing Pathology Workload Required for a Precision Oncology Molecular Test (POMT)

Article Type
Changed
Thu, 12/15/2022 - 14:27

Background

Precision oncology has made nextgeneration sequencing a part of daily practice. With indications for comprehensive genomic profiling expanding, there will be further attendant increases in pathology workload. The pathology workforce shortage is one of the greatest barriers to precision oncology and an understanding of pathology workload associated with POMTs is necessary to address this barrier and plan for the future.

Methods

In this presentation we aim to provide, or at least contribute to, such an understanding through a review of the process at our site and measurement of associated time for each step. We began by conceptualizing the process in order to develop a process map. We then measured the average time for each step. We reviewed our anatomic pathology records for 2021 to determine the number of POMTs then calculated cumulative time investment on POMTs. A theoretical number of relative value units (RVUs) for POMTs was calculated using the new pathology clinical consultation CPT codes (80503-80506), and this was compared to the total anatomic pathology RVUs actually generated in 2021.

Results

Of the 7007 anatomic pathology cases, there were 706 cancers and 446 that required POMTs. At our institution, it was determined that on average 1.5 hours – about 50 minutes of pathologist time and 40 minutes of technician time – was needed to complete the tasks necessary to fulfillment of requests for POMTs. For all of 2021, 669 hours of pathology staff time were dedicated to POMTs. With the ability to bill for this time, we would have generated 13.2% (1142/8640) more anatomic pathology RVUs.

Conculsions

In light of this, we have implemented measures to bill for these formerly uncaptured activities such that our documented productivity more accurately reflects our workload. This will hopefully result in more appropriate resource allocation such that the barrier created by pathology understaffing is recast as a buttress in support of precision oncology practice.

Issue
Federal Practitioner - 39(4)s
Publications
Topics
Page Number
S18
Sections

Background

Precision oncology has made nextgeneration sequencing a part of daily practice. With indications for comprehensive genomic profiling expanding, there will be further attendant increases in pathology workload. The pathology workforce shortage is one of the greatest barriers to precision oncology and an understanding of pathology workload associated with POMTs is necessary to address this barrier and plan for the future.

Methods

In this presentation we aim to provide, or at least contribute to, such an understanding through a review of the process at our site and measurement of associated time for each step. We began by conceptualizing the process in order to develop a process map. We then measured the average time for each step. We reviewed our anatomic pathology records for 2021 to determine the number of POMTs then calculated cumulative time investment on POMTs. A theoretical number of relative value units (RVUs) for POMTs was calculated using the new pathology clinical consultation CPT codes (80503-80506), and this was compared to the total anatomic pathology RVUs actually generated in 2021.

Results

Of the 7007 anatomic pathology cases, there were 706 cancers and 446 that required POMTs. At our institution, it was determined that on average 1.5 hours – about 50 minutes of pathologist time and 40 minutes of technician time – was needed to complete the tasks necessary to fulfillment of requests for POMTs. For all of 2021, 669 hours of pathology staff time were dedicated to POMTs. With the ability to bill for this time, we would have generated 13.2% (1142/8640) more anatomic pathology RVUs.

Conculsions

In light of this, we have implemented measures to bill for these formerly uncaptured activities such that our documented productivity more accurately reflects our workload. This will hopefully result in more appropriate resource allocation such that the barrier created by pathology understaffing is recast as a buttress in support of precision oncology practice.

Background

Precision oncology has made nextgeneration sequencing a part of daily practice. With indications for comprehensive genomic profiling expanding, there will be further attendant increases in pathology workload. The pathology workforce shortage is one of the greatest barriers to precision oncology and an understanding of pathology workload associated with POMTs is necessary to address this barrier and plan for the future.

Methods

In this presentation we aim to provide, or at least contribute to, such an understanding through a review of the process at our site and measurement of associated time for each step. We began by conceptualizing the process in order to develop a process map. We then measured the average time for each step. We reviewed our anatomic pathology records for 2021 to determine the number of POMTs then calculated cumulative time investment on POMTs. A theoretical number of relative value units (RVUs) for POMTs was calculated using the new pathology clinical consultation CPT codes (80503-80506), and this was compared to the total anatomic pathology RVUs actually generated in 2021.

Results

Of the 7007 anatomic pathology cases, there were 706 cancers and 446 that required POMTs. At our institution, it was determined that on average 1.5 hours – about 50 minutes of pathologist time and 40 minutes of technician time – was needed to complete the tasks necessary to fulfillment of requests for POMTs. For all of 2021, 669 hours of pathology staff time were dedicated to POMTs. With the ability to bill for this time, we would have generated 13.2% (1142/8640) more anatomic pathology RVUs.

Conculsions

In light of this, we have implemented measures to bill for these formerly uncaptured activities such that our documented productivity more accurately reflects our workload. This will hopefully result in more appropriate resource allocation such that the barrier created by pathology understaffing is recast as a buttress in support of precision oncology practice.

Issue
Federal Practitioner - 39(4)s
Issue
Federal Practitioner - 39(4)s
Page Number
S18
Page Number
S18
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 08/30/2022 - 15:15
Un-Gate On Date
Tue, 08/30/2022 - 15:15
Use ProPublica
CFC Schedule Remove Status
Tue, 08/30/2022 - 15:15
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

A Rare Case of HHV8+ Multicentric Castleman Disease Presenting as Dermatitis

Article Type
Changed
Tue, 01/31/2023 - 08:30

Introduction

Castleman disease (CD) is a rare non-neoplastic disorder presenting as lymphadenopathy. Skin involvement and progression to lymphomas are uncommon, and such presentation can pose a diagnostic challenge. We describe an interesting case of multicentric CD presenting as a rash.

Case Description

A 79-year-old male presented with a 1-year history of blanchable maculopapular rash and new onset dyspnea in the absence of fever, fatigue or weight loss. Examination revealed axillary, cervical and inguinal lymphadenopathy, and firm splenomegaly. Initial labs were notable for leukocytosis, occasional lymphoplasmacytic cells, anemia, thrombocytopenia, negative HIV screen, and elevated ESR and LDH. Further testing identified polyclonal hypergammaglobulinemia. CT scans revealed generalized lymphadenopathy, splenomegaly with infarcts and unilateral pleural effusion. An inguinal lymph node needle biopsy, skin biopsy and pleural fluid cytology were concerning for lymphoplasmacytic, so he was started on rituximab and bendamustine. However, B cell clonality could not be demonstrated, making these findings concerning for Castleman disease.

Results

Human herpesvirus 8 (HHV-8) testing performed on the inguinal lymph node sample came out positive, and he was diagnosed with HHV-8 positive multicentric Castleman disease and continued on weekly rituximab. He demonstrated an excellent response with complete resolution of rash, palpable lymphadenopathy and anemia after 4 cycles of treatment.

Discussion

Castleman disease (CD) is a rare disorder of polyclonal B cell proliferation classically presenting as lymphadenopathy with constitutional symptoms. Cutaneous presentations include eruptive angiomas or petechial rash but can be variable. Intrinsic or viral IL-6 play a key role in the pathogenesis of the disease. CD can be localised or multicentric (related to HHV-8 +/- HIV or idiopathic), and these subtypes differ in prognosis and management, with HIV and HHV-8 co-positivity indicating worse outcomes. While human IL-6 in unicentric and idiopathic multicentric disease respond well to IL-6 receptor antagonists, viral IL-6 in HHV-8 associated cases has a limited response. This is the rationale for preferring anti-CD20 therapy with rituximab in these patients.

Conculsions

Correct biopsy specimen, keen analysis of distinct pathologic features, and HHV-8 testing on tissue sample guide the diagnosis as HHV-8 serology can be falsely negative.

Issue
Federal Practitioner - 39(4)s
Publications
Topics
Page Number
S17
Sections

Introduction

Castleman disease (CD) is a rare non-neoplastic disorder presenting as lymphadenopathy. Skin involvement and progression to lymphomas are uncommon, and such presentation can pose a diagnostic challenge. We describe an interesting case of multicentric CD presenting as a rash.

Case Description

A 79-year-old male presented with a 1-year history of blanchable maculopapular rash and new onset dyspnea in the absence of fever, fatigue or weight loss. Examination revealed axillary, cervical and inguinal lymphadenopathy, and firm splenomegaly. Initial labs were notable for leukocytosis, occasional lymphoplasmacytic cells, anemia, thrombocytopenia, negative HIV screen, and elevated ESR and LDH. Further testing identified polyclonal hypergammaglobulinemia. CT scans revealed generalized lymphadenopathy, splenomegaly with infarcts and unilateral pleural effusion. An inguinal lymph node needle biopsy, skin biopsy and pleural fluid cytology were concerning for lymphoplasmacytic, so he was started on rituximab and bendamustine. However, B cell clonality could not be demonstrated, making these findings concerning for Castleman disease.

Results

Human herpesvirus 8 (HHV-8) testing performed on the inguinal lymph node sample came out positive, and he was diagnosed with HHV-8 positive multicentric Castleman disease and continued on weekly rituximab. He demonstrated an excellent response with complete resolution of rash, palpable lymphadenopathy and anemia after 4 cycles of treatment.

Discussion

Castleman disease (CD) is a rare disorder of polyclonal B cell proliferation classically presenting as lymphadenopathy with constitutional symptoms. Cutaneous presentations include eruptive angiomas or petechial rash but can be variable. Intrinsic or viral IL-6 play a key role in the pathogenesis of the disease. CD can be localised or multicentric (related to HHV-8 +/- HIV or idiopathic), and these subtypes differ in prognosis and management, with HIV and HHV-8 co-positivity indicating worse outcomes. While human IL-6 in unicentric and idiopathic multicentric disease respond well to IL-6 receptor antagonists, viral IL-6 in HHV-8 associated cases has a limited response. This is the rationale for preferring anti-CD20 therapy with rituximab in these patients.

Conculsions

Correct biopsy specimen, keen analysis of distinct pathologic features, and HHV-8 testing on tissue sample guide the diagnosis as HHV-8 serology can be falsely negative.

Introduction

Castleman disease (CD) is a rare non-neoplastic disorder presenting as lymphadenopathy. Skin involvement and progression to lymphomas are uncommon, and such presentation can pose a diagnostic challenge. We describe an interesting case of multicentric CD presenting as a rash.

Case Description

A 79-year-old male presented with a 1-year history of blanchable maculopapular rash and new onset dyspnea in the absence of fever, fatigue or weight loss. Examination revealed axillary, cervical and inguinal lymphadenopathy, and firm splenomegaly. Initial labs were notable for leukocytosis, occasional lymphoplasmacytic cells, anemia, thrombocytopenia, negative HIV screen, and elevated ESR and LDH. Further testing identified polyclonal hypergammaglobulinemia. CT scans revealed generalized lymphadenopathy, splenomegaly with infarcts and unilateral pleural effusion. An inguinal lymph node needle biopsy, skin biopsy and pleural fluid cytology were concerning for lymphoplasmacytic, so he was started on rituximab and bendamustine. However, B cell clonality could not be demonstrated, making these findings concerning for Castleman disease.

Results

Human herpesvirus 8 (HHV-8) testing performed on the inguinal lymph node sample came out positive, and he was diagnosed with HHV-8 positive multicentric Castleman disease and continued on weekly rituximab. He demonstrated an excellent response with complete resolution of rash, palpable lymphadenopathy and anemia after 4 cycles of treatment.

Discussion

Castleman disease (CD) is a rare disorder of polyclonal B cell proliferation classically presenting as lymphadenopathy with constitutional symptoms. Cutaneous presentations include eruptive angiomas or petechial rash but can be variable. Intrinsic or viral IL-6 play a key role in the pathogenesis of the disease. CD can be localised or multicentric (related to HHV-8 +/- HIV or idiopathic), and these subtypes differ in prognosis and management, with HIV and HHV-8 co-positivity indicating worse outcomes. While human IL-6 in unicentric and idiopathic multicentric disease respond well to IL-6 receptor antagonists, viral IL-6 in HHV-8 associated cases has a limited response. This is the rationale for preferring anti-CD20 therapy with rituximab in these patients.

Conculsions

Correct biopsy specimen, keen analysis of distinct pathologic features, and HHV-8 testing on tissue sample guide the diagnosis as HHV-8 serology can be falsely negative.

Issue
Federal Practitioner - 39(4)s
Issue
Federal Practitioner - 39(4)s
Page Number
S17
Page Number
S17
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 08/30/2022 - 15:00
Un-Gate On Date
Tue, 08/30/2022 - 15:00
Use ProPublica
CFC Schedule Remove Status
Tue, 08/30/2022 - 15:00
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

Identification of Clinically Actionable Genomic Alterations in Colorectal Cancer Patients From the VA National Precision Oncology Program (NPOP)

Article Type
Changed
Thu, 12/15/2022 - 14:27

Purpose

Colorectal cancer (CRC) is the fourth most common cancer at VA and the third leading cause of cancer-related death in the USA. The VA National Precision Oncology Program (NPOP) was established in 2016 with the goal of implementing standardized, streamlined methods for molecular testing of veterans with cancer and has enabled comprehensive genomic profiling (CGP) and precision medicine as part of routine cancer care. Obtaining CGP of predictive biomarkers in cancer tissue, including mutations in genes (e.g., KRAS, NRAS and BRAF), tumor mutation burden (TMB) and microsatellite instability status (MSI) can be used to support treatment decisions with targeted and immunotherapies.

Methods

In this study we describe the frequencies of these clinical biomarkers in colon adenocarcinoma (COAD), rectal adenocarcinoma (READ), and other colon or rectum histologies (CROT); and compare these frequencies to a published cohort of metastatic CRC using Chi-square test (Yaeger et al., 2018).

Results

A total of 1802 patients with CRC were included in this study. COAD was the most frequent disease site (76.9%) followed by READ (19.1%). Approximately 52.9% of COAD patients harbored at least one highly actionable biomarker (defined as having an FDA-approved indication) including NRAS/ KRAS/BRAF wildtype (38.0%), TMB-H (12.9%), BRAF V600E (9.7%), MSI-H (8.9%), and NTRK fusion or rearrangement (0.3%). About 52.0% of patients with READ had these biomarkers, while this rate was (16.4%) in CROT. Among patients with COAD and READ, those with BRAF V600E mutations were more likely to be older, White, not Hispanic or Latino, and lived in urban areas compared to those without BRAF V600E. Relative to those with NRAS/KRAS/BRAF mutations, patients with NRAS/KRAS/BRAF wildtype were frequently younger. Relative to the frequency of biomarkers from a cBioPortal cohort of metastatic CRC, the frequency of NRAS wildtype was significantly lower in patients with COAD and READ tested through NPOP. 

Consulsions

In this cohort, ~53 % of patients with COAD and 52% of patients with READ have highly actionable biomarkers and are potentially eligible for FDAapproved targeted therapies. Future studies examining cancer outcomes with regard to the use of targeted therapies in the setting of actionable gene alterations, TMB, and MSI are warranted.

Issue
Federal Practitioner - 39(4)s
Publications
Topics
Sections

Purpose

Colorectal cancer (CRC) is the fourth most common cancer at VA and the third leading cause of cancer-related death in the USA. The VA National Precision Oncology Program (NPOP) was established in 2016 with the goal of implementing standardized, streamlined methods for molecular testing of veterans with cancer and has enabled comprehensive genomic profiling (CGP) and precision medicine as part of routine cancer care. Obtaining CGP of predictive biomarkers in cancer tissue, including mutations in genes (e.g., KRAS, NRAS and BRAF), tumor mutation burden (TMB) and microsatellite instability status (MSI) can be used to support treatment decisions with targeted and immunotherapies.

Methods

In this study we describe the frequencies of these clinical biomarkers in colon adenocarcinoma (COAD), rectal adenocarcinoma (READ), and other colon or rectum histologies (CROT); and compare these frequencies to a published cohort of metastatic CRC using Chi-square test (Yaeger et al., 2018).

Results

A total of 1802 patients with CRC were included in this study. COAD was the most frequent disease site (76.9%) followed by READ (19.1%). Approximately 52.9% of COAD patients harbored at least one highly actionable biomarker (defined as having an FDA-approved indication) including NRAS/ KRAS/BRAF wildtype (38.0%), TMB-H (12.9%), BRAF V600E (9.7%), MSI-H (8.9%), and NTRK fusion or rearrangement (0.3%). About 52.0% of patients with READ had these biomarkers, while this rate was (16.4%) in CROT. Among patients with COAD and READ, those with BRAF V600E mutations were more likely to be older, White, not Hispanic or Latino, and lived in urban areas compared to those without BRAF V600E. Relative to those with NRAS/KRAS/BRAF mutations, patients with NRAS/KRAS/BRAF wildtype were frequently younger. Relative to the frequency of biomarkers from a cBioPortal cohort of metastatic CRC, the frequency of NRAS wildtype was significantly lower in patients with COAD and READ tested through NPOP. 

Consulsions

In this cohort, ~53 % of patients with COAD and 52% of patients with READ have highly actionable biomarkers and are potentially eligible for FDAapproved targeted therapies. Future studies examining cancer outcomes with regard to the use of targeted therapies in the setting of actionable gene alterations, TMB, and MSI are warranted.

Purpose

Colorectal cancer (CRC) is the fourth most common cancer at VA and the third leading cause of cancer-related death in the USA. The VA National Precision Oncology Program (NPOP) was established in 2016 with the goal of implementing standardized, streamlined methods for molecular testing of veterans with cancer and has enabled comprehensive genomic profiling (CGP) and precision medicine as part of routine cancer care. Obtaining CGP of predictive biomarkers in cancer tissue, including mutations in genes (e.g., KRAS, NRAS and BRAF), tumor mutation burden (TMB) and microsatellite instability status (MSI) can be used to support treatment decisions with targeted and immunotherapies.

Methods

In this study we describe the frequencies of these clinical biomarkers in colon adenocarcinoma (COAD), rectal adenocarcinoma (READ), and other colon or rectum histologies (CROT); and compare these frequencies to a published cohort of metastatic CRC using Chi-square test (Yaeger et al., 2018).

Results

A total of 1802 patients with CRC were included in this study. COAD was the most frequent disease site (76.9%) followed by READ (19.1%). Approximately 52.9% of COAD patients harbored at least one highly actionable biomarker (defined as having an FDA-approved indication) including NRAS/ KRAS/BRAF wildtype (38.0%), TMB-H (12.9%), BRAF V600E (9.7%), MSI-H (8.9%), and NTRK fusion or rearrangement (0.3%). About 52.0% of patients with READ had these biomarkers, while this rate was (16.4%) in CROT. Among patients with COAD and READ, those with BRAF V600E mutations were more likely to be older, White, not Hispanic or Latino, and lived in urban areas compared to those without BRAF V600E. Relative to those with NRAS/KRAS/BRAF mutations, patients with NRAS/KRAS/BRAF wildtype were frequently younger. Relative to the frequency of biomarkers from a cBioPortal cohort of metastatic CRC, the frequency of NRAS wildtype was significantly lower in patients with COAD and READ tested through NPOP. 

Consulsions

In this cohort, ~53 % of patients with COAD and 52% of patients with READ have highly actionable biomarkers and are potentially eligible for FDAapproved targeted therapies. Future studies examining cancer outcomes with regard to the use of targeted therapies in the setting of actionable gene alterations, TMB, and MSI are warranted.

Issue
Federal Practitioner - 39(4)s
Issue
Federal Practitioner - 39(4)s
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 08/30/2022 - 15:00
Un-Gate On Date
Tue, 08/30/2022 - 15:00
Use ProPublica
CFC Schedule Remove Status
Tue, 08/30/2022 - 15:00
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

Evaluation of the Prostate Cancer Molecular Testing Pathway (PCMTP) Within the Veterans Health Administration (VHA)

Article Type
Changed
Thu, 12/15/2022 - 14:27

Purpose

The PCMTP was developed to provide standardized decision support for molecular testing for veterans with prostate cancer.

Background

Prior to the precision medicine era, molecular tumor testing in prostate cancer was not standard of care. Field practitioners were unfamiliar with the role of molecular testing in clinical care. The PCMTP provides direction for germline and tumor testing in appropriate patients with prostate cancer. The expectation is that at least 80% of veterans will be pathway adherent. The PCMTP is an Oncology Clinical Pathway (OCP) that supports evidence-based practice providing highquality, safe, and cost-effective care for veterans reducing variability of care in the VHA.

Methods

The National Oncology Program Office assembled a Prostate Cancer Team (PCT) to develop OCPs. The pathways were incorporated into note templates that record clinical decisions using text and metadata (Health Factors [HF]), and record pathway adherence for the 4 key nodes of the PCMTP. The templates were pilot-tested and improved using an iterative process over a 3-month period. Further evaluation was conducted by the Office of Human Factors Engineering and the National Clinical Template Workgroup, utilizing a heuristic evaluation to ensure standardization, interoperability, and reduce duplication. HF data were retrieved from the Corporate Data Warehouse using a custom-built dashboard. Descriptive statistics of PCMTP use are presented.

Results

Between 4/1/2021 and 6/22/2022, 6276 health factors were generated from 1707 unique veterans in whom this clinical pathway was accessed. 328 distinct providers participated at 61 sites. Average veteran age was 73 years. (range 45-100) including 42% Black and 56% White. Of 1243 veterans considered for germline testing, 96.6% had germline testing ordered and for 1102 veterans considered for tumor testing, 93.3% had tumor testing ordered.

Conclusions

Pathway adherence exceeded the 80% benchmark. Race representation was diverse and reflective of the VA prostate cancer population. About 46% of VA oncology practices have used the PCMTP for ~11% of the estimated 15,000 veterans with metastatic prostate cancer in VHA. Increased use of this pathway is expected to improve outcomes for veterans with prostate cancer

Issue
Federal Practitioner - 39(4)s
Publications
Topics
Sections

Purpose

The PCMTP was developed to provide standardized decision support for molecular testing for veterans with prostate cancer.

Background

Prior to the precision medicine era, molecular tumor testing in prostate cancer was not standard of care. Field practitioners were unfamiliar with the role of molecular testing in clinical care. The PCMTP provides direction for germline and tumor testing in appropriate patients with prostate cancer. The expectation is that at least 80% of veterans will be pathway adherent. The PCMTP is an Oncology Clinical Pathway (OCP) that supports evidence-based practice providing highquality, safe, and cost-effective care for veterans reducing variability of care in the VHA.

Methods

The National Oncology Program Office assembled a Prostate Cancer Team (PCT) to develop OCPs. The pathways were incorporated into note templates that record clinical decisions using text and metadata (Health Factors [HF]), and record pathway adherence for the 4 key nodes of the PCMTP. The templates were pilot-tested and improved using an iterative process over a 3-month period. Further evaluation was conducted by the Office of Human Factors Engineering and the National Clinical Template Workgroup, utilizing a heuristic evaluation to ensure standardization, interoperability, and reduce duplication. HF data were retrieved from the Corporate Data Warehouse using a custom-built dashboard. Descriptive statistics of PCMTP use are presented.

Results

Between 4/1/2021 and 6/22/2022, 6276 health factors were generated from 1707 unique veterans in whom this clinical pathway was accessed. 328 distinct providers participated at 61 sites. Average veteran age was 73 years. (range 45-100) including 42% Black and 56% White. Of 1243 veterans considered for germline testing, 96.6% had germline testing ordered and for 1102 veterans considered for tumor testing, 93.3% had tumor testing ordered.

Conclusions

Pathway adherence exceeded the 80% benchmark. Race representation was diverse and reflective of the VA prostate cancer population. About 46% of VA oncology practices have used the PCMTP for ~11% of the estimated 15,000 veterans with metastatic prostate cancer in VHA. Increased use of this pathway is expected to improve outcomes for veterans with prostate cancer

Purpose

The PCMTP was developed to provide standardized decision support for molecular testing for veterans with prostate cancer.

Background

Prior to the precision medicine era, molecular tumor testing in prostate cancer was not standard of care. Field practitioners were unfamiliar with the role of molecular testing in clinical care. The PCMTP provides direction for germline and tumor testing in appropriate patients with prostate cancer. The expectation is that at least 80% of veterans will be pathway adherent. The PCMTP is an Oncology Clinical Pathway (OCP) that supports evidence-based practice providing highquality, safe, and cost-effective care for veterans reducing variability of care in the VHA.

Methods

The National Oncology Program Office assembled a Prostate Cancer Team (PCT) to develop OCPs. The pathways were incorporated into note templates that record clinical decisions using text and metadata (Health Factors [HF]), and record pathway adherence for the 4 key nodes of the PCMTP. The templates were pilot-tested and improved using an iterative process over a 3-month period. Further evaluation was conducted by the Office of Human Factors Engineering and the National Clinical Template Workgroup, utilizing a heuristic evaluation to ensure standardization, interoperability, and reduce duplication. HF data were retrieved from the Corporate Data Warehouse using a custom-built dashboard. Descriptive statistics of PCMTP use are presented.

Results

Between 4/1/2021 and 6/22/2022, 6276 health factors were generated from 1707 unique veterans in whom this clinical pathway was accessed. 328 distinct providers participated at 61 sites. Average veteran age was 73 years. (range 45-100) including 42% Black and 56% White. Of 1243 veterans considered for germline testing, 96.6% had germline testing ordered and for 1102 veterans considered for tumor testing, 93.3% had tumor testing ordered.

Conclusions

Pathway adherence exceeded the 80% benchmark. Race representation was diverse and reflective of the VA prostate cancer population. About 46% of VA oncology practices have used the PCMTP for ~11% of the estimated 15,000 veterans with metastatic prostate cancer in VHA. Increased use of this pathway is expected to improve outcomes for veterans with prostate cancer

Issue
Federal Practitioner - 39(4)s
Issue
Federal Practitioner - 39(4)s
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 08/30/2022 - 14:45
Un-Gate On Date
Tue, 08/30/2022 - 14:45
Use ProPublica
CFC Schedule Remove Status
Tue, 08/30/2022 - 14:45
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

New Delivery Models Improve Access to Germline Testing for Patients With Advanced Prostate Cancer

Article Type
Changed
Thu, 12/15/2022 - 14:27

Objectives

The VA Oncology Clinical Pathway for Prostate Cancer is the first to include both tumor and germline testing to inform treatment and clinical trial eligibility for advanced disease. Anticipating increased germline testing demand, new germline testing delivery models were created to augment the existing traditional model of referring patients to genetics providers (VA or non-VA) for germline testing. The new models include: a non-traditional model where oncology clinicians perform all pre- and post-test activities and consult genetics when needed, and a hybrid model where oncology clinicians obtain informed consent and place e-consults for germline test ordering, results disclosure, and genetics follow-up, as needed. We sought to assess germline testing by delivery model.

Methods

Data sources included the National Precision Oncology Program (NPOP) dashboard and NPOP-contracted germline testing laboratories. Patient inclusion criteria: living as of 5/2/2021 with VA oncology or urology visits after 5/2/2021. We used multivariate regression to assess associations between patient characteristics and germline testing between 5/3/2021 (pathway launch) and 5/2/2022, accounting for clustering of patients within ordering clinicians.

Results

We identified 16,041 patients from 129 VA facilities with average age 75 years (SD, 8.2; range, 36- 102), 28.7% Black and 60.0% White. Only 5.6% had germline testing ordered by 60 clinicians at 67 facilities with 52.2% of orders by the hybrid model, 32.1% the non-traditional model, and 15.4% the traditional model. Patient characteristics positively associated with germline testing included care at hybrid model (OR, 6.03; 95% CI, 4.62-7.88) or non-traditional model facilities (OR, 5.66; 95% CI, 4.24-7.56) compared to the traditional model, completing tumor molecular testing (OR, 5.80; 95%CI, 4.98-6.75), and Black compared with White race (OR, 1.24; 95%CI, 1.06-1.45). Compared to patients aged < 66 years, patients aged 66-75 years and 76-85 years were less likely to have germline testing (OR, 0.74; 95%CI, 0.60-0.90; and OR, 0.67; 95%CI, 0.53-0.84, respectively).

Conclusions/Implications

Though only a small percentage of patients with advanced prostate cancer had NPOP-supported germline testing since the pathway launch, the new delivery models were instrumental to improving access to germline testing. Ongoing evaluation will help to understand observed demographic differences in germline testing. Implementation and evaluation of strategies that promote adoption of the new germline testing delivery models is needed. 0922FED AVAHO_Abstracts.indd 15 8

Issue
Federal Practitioner - 39(4)s
Publications
Topics
Page Number
S15
Sections

Objectives

The VA Oncology Clinical Pathway for Prostate Cancer is the first to include both tumor and germline testing to inform treatment and clinical trial eligibility for advanced disease. Anticipating increased germline testing demand, new germline testing delivery models were created to augment the existing traditional model of referring patients to genetics providers (VA or non-VA) for germline testing. The new models include: a non-traditional model where oncology clinicians perform all pre- and post-test activities and consult genetics when needed, and a hybrid model where oncology clinicians obtain informed consent and place e-consults for germline test ordering, results disclosure, and genetics follow-up, as needed. We sought to assess germline testing by delivery model.

Methods

Data sources included the National Precision Oncology Program (NPOP) dashboard and NPOP-contracted germline testing laboratories. Patient inclusion criteria: living as of 5/2/2021 with VA oncology or urology visits after 5/2/2021. We used multivariate regression to assess associations between patient characteristics and germline testing between 5/3/2021 (pathway launch) and 5/2/2022, accounting for clustering of patients within ordering clinicians.

Results

We identified 16,041 patients from 129 VA facilities with average age 75 years (SD, 8.2; range, 36- 102), 28.7% Black and 60.0% White. Only 5.6% had germline testing ordered by 60 clinicians at 67 facilities with 52.2% of orders by the hybrid model, 32.1% the non-traditional model, and 15.4% the traditional model. Patient characteristics positively associated with germline testing included care at hybrid model (OR, 6.03; 95% CI, 4.62-7.88) or non-traditional model facilities (OR, 5.66; 95% CI, 4.24-7.56) compared to the traditional model, completing tumor molecular testing (OR, 5.80; 95%CI, 4.98-6.75), and Black compared with White race (OR, 1.24; 95%CI, 1.06-1.45). Compared to patients aged < 66 years, patients aged 66-75 years and 76-85 years were less likely to have germline testing (OR, 0.74; 95%CI, 0.60-0.90; and OR, 0.67; 95%CI, 0.53-0.84, respectively).

Conclusions/Implications

Though only a small percentage of patients with advanced prostate cancer had NPOP-supported germline testing since the pathway launch, the new delivery models were instrumental to improving access to germline testing. Ongoing evaluation will help to understand observed demographic differences in germline testing. Implementation and evaluation of strategies that promote adoption of the new germline testing delivery models is needed. 0922FED AVAHO_Abstracts.indd 15 8

Objectives

The VA Oncology Clinical Pathway for Prostate Cancer is the first to include both tumor and germline testing to inform treatment and clinical trial eligibility for advanced disease. Anticipating increased germline testing demand, new germline testing delivery models were created to augment the existing traditional model of referring patients to genetics providers (VA or non-VA) for germline testing. The new models include: a non-traditional model where oncology clinicians perform all pre- and post-test activities and consult genetics when needed, and a hybrid model where oncology clinicians obtain informed consent and place e-consults for germline test ordering, results disclosure, and genetics follow-up, as needed. We sought to assess germline testing by delivery model.

Methods

Data sources included the National Precision Oncology Program (NPOP) dashboard and NPOP-contracted germline testing laboratories. Patient inclusion criteria: living as of 5/2/2021 with VA oncology or urology visits after 5/2/2021. We used multivariate regression to assess associations between patient characteristics and germline testing between 5/3/2021 (pathway launch) and 5/2/2022, accounting for clustering of patients within ordering clinicians.

Results

We identified 16,041 patients from 129 VA facilities with average age 75 years (SD, 8.2; range, 36- 102), 28.7% Black and 60.0% White. Only 5.6% had germline testing ordered by 60 clinicians at 67 facilities with 52.2% of orders by the hybrid model, 32.1% the non-traditional model, and 15.4% the traditional model. Patient characteristics positively associated with germline testing included care at hybrid model (OR, 6.03; 95% CI, 4.62-7.88) or non-traditional model facilities (OR, 5.66; 95% CI, 4.24-7.56) compared to the traditional model, completing tumor molecular testing (OR, 5.80; 95%CI, 4.98-6.75), and Black compared with White race (OR, 1.24; 95%CI, 1.06-1.45). Compared to patients aged < 66 years, patients aged 66-75 years and 76-85 years were less likely to have germline testing (OR, 0.74; 95%CI, 0.60-0.90; and OR, 0.67; 95%CI, 0.53-0.84, respectively).

Conclusions/Implications

Though only a small percentage of patients with advanced prostate cancer had NPOP-supported germline testing since the pathway launch, the new delivery models were instrumental to improving access to germline testing. Ongoing evaluation will help to understand observed demographic differences in germline testing. Implementation and evaluation of strategies that promote adoption of the new germline testing delivery models is needed. 0922FED AVAHO_Abstracts.indd 15 8

Issue
Federal Practitioner - 39(4)s
Issue
Federal Practitioner - 39(4)s
Page Number
S15
Page Number
S15
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 08/30/2022 - 14:45
Un-Gate On Date
Tue, 08/30/2022 - 14:45
Use ProPublica
CFC Schedule Remove Status
Tue, 08/30/2022 - 14:45
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