Fresh look at ISCHEMIA bolsters conservative message in stable CAD

Article Type
Changed
Tue, 05/11/2021 - 15:44

 

The more complicated a primary endpoint, the greater a puzzle it can be for clinicians to interpret the results. It’s likely even tougher for patients, who don’t help choose the events studied in clinical trials yet are increasingly sharing in the management decisions they influence.

That creates an opening for a more patient-centered take on one of cardiology’s most influential recent studies, ISCHEMIA, which bolsters the case for conservative, med-oriented management over a more invasive initial strategy for patients with stable coronary artery disease (CAD) and positive stress tests, researchers said.

The new, prespecified analysis replaced the trial’s conventional primary endpoint of major adverse cardiac events (MACE) with one based on “days alive out of hospital” (DAOH) and found an early advantage for the conservative approach, with caveats.

Those assigned to the conservative arm benefited with more out-of-hospital days throughout the next 2 years than those in the invasive-management group, owing to the latter’s protocol-mandated early cath-lab work-up with possible revascularization. The difference averaged more than 6 days for much of that time.

But DAOH evened out for the two groups by the fourth year in the analysis of more than 5,000 patients.

Protocol-determined cath procedures accounted for 61% of hospitalizations in the invasively managed group. A secondary DAOH analysis that excluded such required hospital days, also prespecified, showed no meaningful difference between the two strategies over the 4 years, noted the report published online May 3 in JAMA Cardiology.
 

DOAH is ‘very, very important’

The DAOH metric has been a far less common consideration in clinical trials, compared with clinical events, yet in some ways it is as “hard” a metric as mortality, encompasses a broader range of outcomes, and may matter more to patients, it’s been argued.

“The thing patients most value is time at home. So they don’t want to be in the hospital, they don’t want to be away from friends, they want to do recreation, or they may want to work,” lead author Harvey D. White, DSc, Green Lane Cardiovascular Services, Auckland (New Zealand) City Hospital, University of Auckland, told this news organization.

“When we need to talk to patients – and we do need to talk to patients – to have a days-out-of-hospital metric is very, very important,” he said. It is not only patient focused, it’s “meaningful in terms of the seriousness of events,” in that length of hospitalization tracks with clinical severity, observed Dr. White, who is slated to present the analysis May 17 during the virtual American College of Cardiology 2021 scientific sessions.

As previously reported, ISCHEMIA showed no significant effect on the primary endpoint of cardiovascular mortality, MI, or hospitalization for unstable anginaheart failure, or resuscitated cardiac arrest by assignment group over a median 3.2 years. Angina and quality of life measures were improved for patients in the invasive arm.

With an invasive initial strategy, “What we know now is that you get nothing of an advantage in terms of the composite endpoint, and you’re going to spend 6 days more in the hospital in the first 2 years, for largely no benefit,” Dr. White said.

That outlook may apply out to 4 years, the analysis suggests, but could conceivably change if DAOH is reassessed later as the ISCHEMIA follow-up continues for what is now a planned total of 10 years.

Meanwhile, the current findings could enhance doctor-patient discussions about the trade-offs between the two strategies for individuals whose considerations will vary.

“This is a very helpful measure to understand the burden of an approach to the patient,” observed E. Magnus Ohman, MD, an interventional cardiologist at Duke University, Durham, N.C., who was not involved in the trial.

With DAOH as an endpoint, “you as a clinician get another aspect of understanding of a treatment’s impact on a multitude of endpoints.” Days out of hospital, he noted, encompasses the effects of clinical events that often go into composite clinical endpoints – not death, but including nonfatal MI, stroke, need for revascularization, and cardiovascular hospitalization.

To patients with stable CAD who ask whether the invasive approach has merits in their case, the DAOH finding “helps you to say, well, at the end of the day, you will probably be spending an equal amount of time in the hospital. Your price up front is a little bit higher, but over time, the group who gets conservative treatment will catch up.”

The DAOH outcome also avoids the limitations of an endpoint based on time to first event, “not the least of which,” said Dr. White, is that it counts only the first of what might be multiple events of varying clinical impact. Misleadingly, “you can have an event that’s a small troponin rise, but that becomes more important in a person than dying the next day.”

The DAOH analysis was based on 5,179 patients from 37 countries who averaged 64 years of age and of whom 23% were women. The endpoint considered only overnight stays in hospitals, skilled nursing facilities, rehabilitation centers, and nursing homes.

There were many more hospital or extended care facility stays overall in the invasive-management group, 4,002 versus 1,897 for those following the conservative strategy (P < .001), but the numbers flipped after excluding protocol-assigned procedures: 1,568 stays in the invasive group, compared with 1,897 (P = .001)

There were no associations between DAOH and Seattle Angina Questionnaire 7–Angina Frequency scores or DAOH interactions by age, sex, geographic region, or whether the patient had diabetes, prior MI, or heart failure, the report notes.

The primary ISCHEMIA analysis hinted at a possible long-term advantage for the invasive initial strategy in that event curves for the two arms crossed after 2-3 years, Dr. Ohman observed.

Based on that, for younger patients with stable CAD and ischemia at stress testing, “an investment of more hospital days early on might be worth it in the long run.” But ISCHEMIA, he said, “only suggests it, it doesn’t confirm it.”

The study was supported in part by grants from Arbor Pharmaceuticals and AstraZeneca. Devices or medications were provided by Abbott Vascular, Amgen, Arbor, AstraZeneca, Esperion, Medtronic, Merck Sharp & Dohme, Phillips, Omron Healthcare, and Sunovion. Dr. White disclosed receiving grants paid to his institution and fees for serving on a steering committee from Sanofi-Aventis, Regeneron, Eli Lilly, Omthera, American Regent, Eisai, DalCor, CSL Behring, Sanofi-Aventis Australia, and Esperion Therapeutics, and personal fees from Genentech and AstraZeneca. Dr. Ohman reported receiving grants from Abiomed and Cheisi USA, and consulting for Abiomed, Cara Therapeutics, Chiesi USA, Cytokinetics, Imbria Pharmaceuticals, Otsuka Pharmaceuticals, Milestone Pharmaceuticals, and XyloCor Therapeutics.
 

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

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

 

The more complicated a primary endpoint, the greater a puzzle it can be for clinicians to interpret the results. It’s likely even tougher for patients, who don’t help choose the events studied in clinical trials yet are increasingly sharing in the management decisions they influence.

That creates an opening for a more patient-centered take on one of cardiology’s most influential recent studies, ISCHEMIA, which bolsters the case for conservative, med-oriented management over a more invasive initial strategy for patients with stable coronary artery disease (CAD) and positive stress tests, researchers said.

The new, prespecified analysis replaced the trial’s conventional primary endpoint of major adverse cardiac events (MACE) with one based on “days alive out of hospital” (DAOH) and found an early advantage for the conservative approach, with caveats.

Those assigned to the conservative arm benefited with more out-of-hospital days throughout the next 2 years than those in the invasive-management group, owing to the latter’s protocol-mandated early cath-lab work-up with possible revascularization. The difference averaged more than 6 days for much of that time.

But DAOH evened out for the two groups by the fourth year in the analysis of more than 5,000 patients.

Protocol-determined cath procedures accounted for 61% of hospitalizations in the invasively managed group. A secondary DAOH analysis that excluded such required hospital days, also prespecified, showed no meaningful difference between the two strategies over the 4 years, noted the report published online May 3 in JAMA Cardiology.
 

DOAH is ‘very, very important’

The DAOH metric has been a far less common consideration in clinical trials, compared with clinical events, yet in some ways it is as “hard” a metric as mortality, encompasses a broader range of outcomes, and may matter more to patients, it’s been argued.

“The thing patients most value is time at home. So they don’t want to be in the hospital, they don’t want to be away from friends, they want to do recreation, or they may want to work,” lead author Harvey D. White, DSc, Green Lane Cardiovascular Services, Auckland (New Zealand) City Hospital, University of Auckland, told this news organization.

“When we need to talk to patients – and we do need to talk to patients – to have a days-out-of-hospital metric is very, very important,” he said. It is not only patient focused, it’s “meaningful in terms of the seriousness of events,” in that length of hospitalization tracks with clinical severity, observed Dr. White, who is slated to present the analysis May 17 during the virtual American College of Cardiology 2021 scientific sessions.

As previously reported, ISCHEMIA showed no significant effect on the primary endpoint of cardiovascular mortality, MI, or hospitalization for unstable anginaheart failure, or resuscitated cardiac arrest by assignment group over a median 3.2 years. Angina and quality of life measures were improved for patients in the invasive arm.

With an invasive initial strategy, “What we know now is that you get nothing of an advantage in terms of the composite endpoint, and you’re going to spend 6 days more in the hospital in the first 2 years, for largely no benefit,” Dr. White said.

That outlook may apply out to 4 years, the analysis suggests, but could conceivably change if DAOH is reassessed later as the ISCHEMIA follow-up continues for what is now a planned total of 10 years.

Meanwhile, the current findings could enhance doctor-patient discussions about the trade-offs between the two strategies for individuals whose considerations will vary.

“This is a very helpful measure to understand the burden of an approach to the patient,” observed E. Magnus Ohman, MD, an interventional cardiologist at Duke University, Durham, N.C., who was not involved in the trial.

With DAOH as an endpoint, “you as a clinician get another aspect of understanding of a treatment’s impact on a multitude of endpoints.” Days out of hospital, he noted, encompasses the effects of clinical events that often go into composite clinical endpoints – not death, but including nonfatal MI, stroke, need for revascularization, and cardiovascular hospitalization.

To patients with stable CAD who ask whether the invasive approach has merits in their case, the DAOH finding “helps you to say, well, at the end of the day, you will probably be spending an equal amount of time in the hospital. Your price up front is a little bit higher, but over time, the group who gets conservative treatment will catch up.”

The DAOH outcome also avoids the limitations of an endpoint based on time to first event, “not the least of which,” said Dr. White, is that it counts only the first of what might be multiple events of varying clinical impact. Misleadingly, “you can have an event that’s a small troponin rise, but that becomes more important in a person than dying the next day.”

The DAOH analysis was based on 5,179 patients from 37 countries who averaged 64 years of age and of whom 23% were women. The endpoint considered only overnight stays in hospitals, skilled nursing facilities, rehabilitation centers, and nursing homes.

There were many more hospital or extended care facility stays overall in the invasive-management group, 4,002 versus 1,897 for those following the conservative strategy (P < .001), but the numbers flipped after excluding protocol-assigned procedures: 1,568 stays in the invasive group, compared with 1,897 (P = .001)

There were no associations between DAOH and Seattle Angina Questionnaire 7–Angina Frequency scores or DAOH interactions by age, sex, geographic region, or whether the patient had diabetes, prior MI, or heart failure, the report notes.

The primary ISCHEMIA analysis hinted at a possible long-term advantage for the invasive initial strategy in that event curves for the two arms crossed after 2-3 years, Dr. Ohman observed.

Based on that, for younger patients with stable CAD and ischemia at stress testing, “an investment of more hospital days early on might be worth it in the long run.” But ISCHEMIA, he said, “only suggests it, it doesn’t confirm it.”

The study was supported in part by grants from Arbor Pharmaceuticals and AstraZeneca. Devices or medications were provided by Abbott Vascular, Amgen, Arbor, AstraZeneca, Esperion, Medtronic, Merck Sharp & Dohme, Phillips, Omron Healthcare, and Sunovion. Dr. White disclosed receiving grants paid to his institution and fees for serving on a steering committee from Sanofi-Aventis, Regeneron, Eli Lilly, Omthera, American Regent, Eisai, DalCor, CSL Behring, Sanofi-Aventis Australia, and Esperion Therapeutics, and personal fees from Genentech and AstraZeneca. Dr. Ohman reported receiving grants from Abiomed and Cheisi USA, and consulting for Abiomed, Cara Therapeutics, Chiesi USA, Cytokinetics, Imbria Pharmaceuticals, Otsuka Pharmaceuticals, Milestone Pharmaceuticals, and XyloCor Therapeutics.
 

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

 

The more complicated a primary endpoint, the greater a puzzle it can be for clinicians to interpret the results. It’s likely even tougher for patients, who don’t help choose the events studied in clinical trials yet are increasingly sharing in the management decisions they influence.

That creates an opening for a more patient-centered take on one of cardiology’s most influential recent studies, ISCHEMIA, which bolsters the case for conservative, med-oriented management over a more invasive initial strategy for patients with stable coronary artery disease (CAD) and positive stress tests, researchers said.

The new, prespecified analysis replaced the trial’s conventional primary endpoint of major adverse cardiac events (MACE) with one based on “days alive out of hospital” (DAOH) and found an early advantage for the conservative approach, with caveats.

Those assigned to the conservative arm benefited with more out-of-hospital days throughout the next 2 years than those in the invasive-management group, owing to the latter’s protocol-mandated early cath-lab work-up with possible revascularization. The difference averaged more than 6 days for much of that time.

But DAOH evened out for the two groups by the fourth year in the analysis of more than 5,000 patients.

Protocol-determined cath procedures accounted for 61% of hospitalizations in the invasively managed group. A secondary DAOH analysis that excluded such required hospital days, also prespecified, showed no meaningful difference between the two strategies over the 4 years, noted the report published online May 3 in JAMA Cardiology.
 

DOAH is ‘very, very important’

The DAOH metric has been a far less common consideration in clinical trials, compared with clinical events, yet in some ways it is as “hard” a metric as mortality, encompasses a broader range of outcomes, and may matter more to patients, it’s been argued.

“The thing patients most value is time at home. So they don’t want to be in the hospital, they don’t want to be away from friends, they want to do recreation, or they may want to work,” lead author Harvey D. White, DSc, Green Lane Cardiovascular Services, Auckland (New Zealand) City Hospital, University of Auckland, told this news organization.

“When we need to talk to patients – and we do need to talk to patients – to have a days-out-of-hospital metric is very, very important,” he said. It is not only patient focused, it’s “meaningful in terms of the seriousness of events,” in that length of hospitalization tracks with clinical severity, observed Dr. White, who is slated to present the analysis May 17 during the virtual American College of Cardiology 2021 scientific sessions.

As previously reported, ISCHEMIA showed no significant effect on the primary endpoint of cardiovascular mortality, MI, or hospitalization for unstable anginaheart failure, or resuscitated cardiac arrest by assignment group over a median 3.2 years. Angina and quality of life measures were improved for patients in the invasive arm.

With an invasive initial strategy, “What we know now is that you get nothing of an advantage in terms of the composite endpoint, and you’re going to spend 6 days more in the hospital in the first 2 years, for largely no benefit,” Dr. White said.

That outlook may apply out to 4 years, the analysis suggests, but could conceivably change if DAOH is reassessed later as the ISCHEMIA follow-up continues for what is now a planned total of 10 years.

Meanwhile, the current findings could enhance doctor-patient discussions about the trade-offs between the two strategies for individuals whose considerations will vary.

“This is a very helpful measure to understand the burden of an approach to the patient,” observed E. Magnus Ohman, MD, an interventional cardiologist at Duke University, Durham, N.C., who was not involved in the trial.

With DAOH as an endpoint, “you as a clinician get another aspect of understanding of a treatment’s impact on a multitude of endpoints.” Days out of hospital, he noted, encompasses the effects of clinical events that often go into composite clinical endpoints – not death, but including nonfatal MI, stroke, need for revascularization, and cardiovascular hospitalization.

To patients with stable CAD who ask whether the invasive approach has merits in their case, the DAOH finding “helps you to say, well, at the end of the day, you will probably be spending an equal amount of time in the hospital. Your price up front is a little bit higher, but over time, the group who gets conservative treatment will catch up.”

The DAOH outcome also avoids the limitations of an endpoint based on time to first event, “not the least of which,” said Dr. White, is that it counts only the first of what might be multiple events of varying clinical impact. Misleadingly, “you can have an event that’s a small troponin rise, but that becomes more important in a person than dying the next day.”

The DAOH analysis was based on 5,179 patients from 37 countries who averaged 64 years of age and of whom 23% were women. The endpoint considered only overnight stays in hospitals, skilled nursing facilities, rehabilitation centers, and nursing homes.

There were many more hospital or extended care facility stays overall in the invasive-management group, 4,002 versus 1,897 for those following the conservative strategy (P < .001), but the numbers flipped after excluding protocol-assigned procedures: 1,568 stays in the invasive group, compared with 1,897 (P = .001)

There were no associations between DAOH and Seattle Angina Questionnaire 7–Angina Frequency scores or DAOH interactions by age, sex, geographic region, or whether the patient had diabetes, prior MI, or heart failure, the report notes.

The primary ISCHEMIA analysis hinted at a possible long-term advantage for the invasive initial strategy in that event curves for the two arms crossed after 2-3 years, Dr. Ohman observed.

Based on that, for younger patients with stable CAD and ischemia at stress testing, “an investment of more hospital days early on might be worth it in the long run.” But ISCHEMIA, he said, “only suggests it, it doesn’t confirm it.”

The study was supported in part by grants from Arbor Pharmaceuticals and AstraZeneca. Devices or medications were provided by Abbott Vascular, Amgen, Arbor, AstraZeneca, Esperion, Medtronic, Merck Sharp & Dohme, Phillips, Omron Healthcare, and Sunovion. Dr. White disclosed receiving grants paid to his institution and fees for serving on a steering committee from Sanofi-Aventis, Regeneron, Eli Lilly, Omthera, American Regent, Eisai, DalCor, CSL Behring, Sanofi-Aventis Australia, and Esperion Therapeutics, and personal fees from Genentech and AstraZeneca. Dr. Ohman reported receiving grants from Abiomed and Cheisi USA, and consulting for Abiomed, Cara Therapeutics, Chiesi USA, Cytokinetics, Imbria Pharmaceuticals, Otsuka Pharmaceuticals, Milestone Pharmaceuticals, and XyloCor Therapeutics.
 

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

Screening High-Risk Women Veterans for Breast Cancer

Article Type
Changed
Wed, 01/04/2023 - 17:17

The number of women seeking care from the Veterans Health Administration (VHA) is increasing.1 In 2015, there were 2 million women veterans in the United States, which is 9.4% of the total veteran population. This group is expected to increase at an average of about 18,000 women per year for the next 10 years.2 The percentage of women veterans who are US Department of Veterans Affairs (VA) users aged 45 to 64 years rose 46% from 2000 to 2015.1,3-4 It is estimated that 15% of veterans who used VA services in 2020 were women.1 Nineteen percent of women veterans are Black.1 The median age of women veterans in 2015 was 50 years.5 Breast cancer is the leading cancer affecting female veterans, and data suggest they have an increased risk of breast cancer based on unique service-related exposures.1,6-9

In the US, about 10 million women are eligible for breast cancer preventive therapy, including, but not limited to, medications, surgery, or lifestyle changes.10 Secondary prevention options include change in surveillance that can reduce their risk or identify cancer at an earlier stage when treatment is more effective. The United States Preventive Services Task Force, the National Comprehensive Cancer Network, the American Society for Clinical Oncology, the National Institute for Health and Care Excellence, and the Oncology Nursing Society recommend screening women aged ≥ 35 years to assess breast cancer risk.11-18 If a woman is at increased risk, she may be a candidate for chemoprevention, prozphylactic surgery, and possibly an enhanced screening regimen.

Urban and minority women are an understudied population. Most veterans (75%) live in urban or suburban settings.19,20 Urban veteran women constitute an important potential study population.

Chemoprevention measures have been underused because of factors involving both women and their health care providers. A large proportion of women are unaware of their higher risk status due to lack of adequate screening and risk assessment.21,22 In addition to patient lack of awareness of their high-risk status, primary care physicians are also reluctant to prescribe chemopreventive agents due to a lack of comfort or familiarity with the risks and benefits.23-26 The STAR2015, BCPT2005, IBIS2014, MAP3 2011, IBIS-I 2014, and IBIS II 2014 studies clearly demonstrate a 49 to 62% reduction in risk for women using chemoprevention such as selective estrogen receptor modulators or aromatase inhibitors, respectively.27-32 Yet only 4 to 9% of high-risk women not enrolled in a clinical trial are using chemoprevention.33-39

The possibility of developing breast cancer also may be increased because of a positive family history or being a member of a family in which there is a known susceptibility gene mutation.40 Based on these risk factors, women may be eligible for tailored follow-up and genetic counseling.41-44

Nationally, 7 to 10% of the civilian US population will experience posttraumatic stress disorder (PTSD).45 The rates are remarkably higher for women veterans, with roughly 20% diagnosed with PTSD.46,47 Anxiety and PTSD have been implicated in poor adherence to medical advice.48,49

In 2014, a national VA multidisciplinary group focused on breast cancer prevention, detection, treatment, and research to address breast health in the growing population of women veterans. High-risk breast cancer screenings are not routinely carried out by the VA in primary care, women’s health, or oncology services. Furthermore, the recording of screening questionnaire results was not synchronized until a standard questionnaire was created and approved as a template by this group in the VA electronic medical record (EMR) in 2015.

Several prediction models can identify which women are at an increased risk of developing breast cancer. The most commonly used risk assessment model, the Gail breast cancer risk assessment tool (BCRAT), has been refined to include women of additional ethnicities (https://www.cancer.gov/bcrisktool).

This pilot project was launched to identify an effective manner to screen women veterans regarding their risk of developing breast cancer and refer them for chemoprevention education or genetic counseling as appropriate.

 

 

Methods

A high-risk breast cancer screening questionnaire based on the Gail BCRAT and including lifestyle questions was developed and included as a note template in the VA EMR. The James J. Peters VA Medical Center, Bronx, NY (JJPVAMC) and the Washington DC VA Medical Center (DCVAMC) ran a pilot study between 2015 and 2018 using this breast cancer screening questionnaire to collect data from women veterans. Quality Executive Committee and institutional review board approvals were granted respectively.

Eligibility criteria included women aged ≥ 35 years with no personal history of breast cancer. Most patients were self-referred, but participants also were recruited during VA Breast Cancer Awareness month events, health fairs, or at informational tables in the hospital lobbies. After completing the 20 multiple choice questionnaire with a study team member, either in person or over the phone, a 5-year and lifetime risk of invasive breast cancer was calculated using the Gail BCRAT. A woman is considered high risk and eligible for chemoprevention if her 5-year risk is > 1.66% or her lifetime risk is ≥ 20%. Eligibility for genetic counseling is based on the Breast Cancer Referral Screening Tool, which includes a personal or family history of breast or ovarian cancer and Jewish ancestry.

All patients were notified of their average or high risk status by a clinician. Those who were deemed to be average risk received a follow-up letter in the mail with instructions (eg, to follow-up with a yearly mammogram). Those who were deemed to be high risk for developing breast cancer were asked to come in for an appointment with the study principal investigator (a VA oncologist/breast cancer specialist) to discuss prevention options, further screening, or referrals to genetic counseling. Depending on a patient’s other health factors, a woman at high risk for developing breast cancer also may be a candidate for chemoprevention with tamoxifen, raloxifene, exemestane, anastrozole, or letrozole.

Data on the participant’s lifestyle, including exercise, diet, and smoking, were evaluated to determine whether these factors had an impact on risk status.

Results

The JJP and DC VAMCs screened 103 women veterans between 2015 and 2018. Four patients were excluded for nonveteran (spousal) status, leaving 99 women veterans with a mean age of 54 years. The most common self-reported races were Black (60%), non-Hispanic White (14%), and Hispanic or Latino (13%) (Table 1).

Women veterans in our study were nearly 3-times more likely than the general population were to receive a high-risk Gail Score/BCRAT (35% vs 13%, respectively).50,51 Of this subset, 46% had breast biopsies, and 86% had a positive family history. Thirty-one percent of Black women in our study were high risk, while nationally, 8.2 to 13.3% of Black women aged 50 to 59 years are considered high risk.50,51 Of the Black high-risk group with a high Gail/BCRAT score, 94% had a positive family history, and 33% had a history of breast biopsy (Table 2).

Of the 35 high-risk patients 26 (74%) patients accepted consultations for chemoprevention and 5 (19%) started chemoprevention. Of this high-risk group, 13 (37%) patients were referred for genetic counseling (Table 3).44 The prevalence of PTSD was present in 31% of high-risk women and 29% of the cohort (Figure).The lifestyle questions indicated that, among all participants, 79% had an overweight or obese body mass index; 58% exercised weekly; 51% consumed alcohol; 14% were smokers; and 21% consumed 3 to 4 servings of fruits/vegetables daily.

 

 

Discussion

Breast cancer is the most common cancer in women.52 The number of women with breast cancer in the VHA has more than tripled from 1995 to 2012.1 The lifetime risk of developing breast cancer in the general population is about 13%.50 This rate can be affected by risk factors including age, hormone exposure, family history, radiation exposure, and lifestyle factors, such as weight and alcohol use.6,52-56 In the United States, invasive breast cancer affects 1 in 8 women.50,52,57

Our screened population showed nearly 3 times as many women veterans were at an increased risk for breast cancer when compared with historical averages in US women. This difference may be based on a high rate of prior breast biopsies or positive family history, although a provocative study using the Surveillance, Epidemiology, and End Results database showed military women to have higher rates of breast cancer as well.9 Historically, Blacks are vastly understudied in clinical research with only 5% representation on a national level.5,58 The urban locations of both pilot sites (Washington, DC and Bronx, NY) allowed for the inclusion of minority patients in our study. We found that the rates of breast cancer in Black women veterans to be higher than seen nationally, possibly prompting further screening initiatives for this understudied population.

Our pilot study’s chemoprevention utilization (19%) was double the < 10% seen in the national population.33-35 The presence of a knowledgeable breast health practitioner to recruit study participants and offer personalized counseling to women veterans is a likely factor in overcoming barriers to chemopreventive acceptance. These participants may have been motivated to seek care for their high-risk status given a strong family history and prior breast biopsies.

Interestingly, a 3-fold higher PTSD rate was seen in this pilot population (29%) when compared with PTSD rates in the general female population (7-10%) and still one-third higher than the general population of women veterans (20%).45-47 Mental health, anxiety, and PTSD have been barriers to patients who sought treatment and have been implicated in poor adherence to medical advice.48,49 Cancer screening can induce anxiety in patients, and it may be amplified in patients with PTSD. It was remarkable that although adherence with screening recommendations is decreased when PTSD is present, our patient population demonstrated a higher rate of screening adherence.

Women who are seen at the VA often use multiple clinical specialties, and their EMR can be accessed across VA medical centers nationwide. Therefore, identifying women veterans who meet screening criteria is easily attainable within the VA.

When comparing high-risk with average risk women, the lifestyle results (BMI, smoking history, exercise and consumption of fruits, vegetables and alcohol) were essentially the same. Lifestyle factors were similar to national population rates and were unlikely to impact risk levels.

Limitations

Study limitations included a high number of self-referrals and the large percentage of patients with a family history of breast cancer, making them more likely to seek screening. The higher-than-average risk of breast cancer may be driven by a high rate of breast biopsies and a strong family history. Lifestyle metrics could not be accurately compared to other national assessments of lifestyle factors due to the difference in data points that we used or the format of our questions.

 

 

Conclusions

As the number of women veterans increases and the incidence of breast cancer in women veterans rise, chemoprevention options should follow national guidelines. To our knowledge, this is the only oncology study with 60% Black women veterans. This study had a higher participation rate for Black women veterans than is typically seen in national research studies and shows the VA to be a germane source for further understanding of an understudied population that may benefit from increased screening for breast cancer.

A team-based, multidisciplinary model that meets the unique healthcare needs of women veterans results in a patient-centric delivery of care for assessing breast cancer risk status and prevention options. This model can be replicated nationally by directing primary care physicians and women’s health practitioners to a risk-assessment questionnaire and referring high-risk women for appropriate preventative care. Given that these results show chemoprevention adherence rates doubled those seen nationally, perhaps techniques used within this VA pilot study may be adapted to decrease breast cancer incidence nationally.

Since the rate of PTSD among women veterans is triple the national average, we would expect adherence rates to be lower in our patient cohort. However, the multidisciplinary approach we used in this study (eg, 1:1 consultation with oncologist; genetic counseling referrals; mental health support available), may have improved adherence rates. Perhaps the high rates of PTSD seen in the VA patient population can be a useful way to explore patient adherence rates in those with mental illness and medical conditions.

Future research with a larger cohort may lead to greater insight into the correlation between PTSD and adherence to treatment. Exploring the connection between breast cancer, epigenetics, and specific military service-related exposures could be an area of analysis among this veteran population exhibiting increased breast cancer rates. VAMCs are situated in rural, suburban, and urban locations across the United States and offers a diverse socioeconomic and ethnic patient population for inclusion in clinical investigations. Women veterans make up a small subpopulation of women in the United States, but it is worth considering VA patients as an untapped resource for research collaboration.

Acknowledgements

The authors thank Steven Sanchez and Marissa Vallette, PhD, Breast Health Research Group. This research project was approved by the James J. Peters VA Medical Center Quality Executive Committee and the Washington, DC VA Medical Center Institutional Review Board. This work was supported by the US Department of Veterans Affairs. This work did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author disclosures

The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

References

1. US Department of Veterans Affairs. National Center for Veterans Analysis and Statistics. The past, present and future of women veterans. Published February 2017. Accessed April 28, 2021. https://www.va.gov/vetdata/docs/specialreports/women_veterans_2015_final.pdf.

2. Frayne SM, Carney DV, Bastian L, et al. The VA Women’s Health Practice-Based Research Network: amplifying women veterans’ voices in VA research. J Gen Intern Med. 2013;28 Suppl 2(Suppl 2):S504-S509. doi:10.1007/s11606-013-2476-3

3. US Department of Veterans Affairs, Veterans Health Administration, Women’s Health Evaluation Initiative, Women Veterans Health Strategic Health Care Group. Sourcebook: women veterans in the Veterans Health Administration. Volume 1: Sociodemographic characteristics and use of VHA care. Published December 2010. Accessed April 12, 2021. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=2455

4. Bean-Mayberry B, Yano EM, Bayliss N, Navratil J, Weisman CS, Scholle SH. Federally funded comprehensive women’s health centers: leading innovation in women’s healthcare delivery. J Womens Health (Larchmt). 2007;16(9):1281-1290. doi:10.1089/jwh.2006.0284

5. US Department of Veterans Affairs. National Center for Veterans Analysis and Statistics.VA utilization profile FY 2016. Published November 2017. Accessed April 12, 2021. https://www.va.gov/vetdata/docs/QuickFacts/VA_Utilization_Profile.PDF

6. Ekenga CC, Parks CG, Sandler DP. Chemical exposures in the workplace and breast cancer risk: a prospective cohort study. Int J Cancer. 2015;137(7):1765-1774. doi:10.1002/ijc.29545

7. Rennix CP, Quinn MM, Amoroso PJ, Eisen EA, Wegman DH. Risk of breast cancer among enlisted Army women occupationally exposed to volatile organic compounds. Am J Ind Med. 2005;48(3):157-167. doi:10.1002/ajim.20201

8. Ritz B. Cancer mortality among workers exposed to chemicals during uranium processing. J Occup Environ Med. 1999;41(7):556-566. doi:10.1097/00043764-199907000-00004

9. Zhu K, Devesa SS, Wu H, et al. Cancer incidence in the U.S. military population: comparison with rates from the SEER program. Cancer Epidemiol Biomarkers Prev. 2009;18(6):1740-1745. doi:10.1158/1055-9965.EPI-09-0041

10. Freedman AN, Yu B, Gail MH, et al. Benefit/risk assessment for breast cancer chemoprevention with raloxifene or tamoxifen for women age 50 years or older [published correction appears in J Clin Oncol. 2013 Nov 10;31(32):4167]. J Clin Oncol. 2011;29(17):2327-2333. doi:10.1200/JCO.2010.33.0258

11. Greene, H. Cancer prevention, screening and early detection. In: Gobel BH, Triest-Robertson S, Vogel WH, eds. Advanced Oncology Nursing Certification Review and Resource Manual. 3rd ed. Oncology Nursing Society; 2016:1-34. https://www.ons.org/sites/default/files/publication_pdfs/2%20ADVPrac%20chapter%201.pdf

12. National Comprehensive Cancer Network. NCCN Breast Cancer Risk Reduction. Version 1.2021 NCCN Clinical Practice Guidelines in Oncology. Updated March 24, 2021 Accessed April 12, 2021. https://www.nccn.org/professionals/physician_gls/pdf/breast_risk.pdf

13. US Preventive Services Task Force. Breast cancer: Medications use to reduce risk. Updated September 3, 2019. Accessed April 12, 2021. https://www.uspreventiveservicestaskforce.org/uspstf/recommendation/breast-cancer-medications-for-risk-reduction

14. Moyer VA; U.S. Preventive Services Task Force. Medications to decrease the risk for breast cancer in women: recommendations from the U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2013;159(10):698-708. doi:10.7326/0003-4819-159-10-201311190-00717

15. Boucher JE. Chemoprevention: an overview of pharmacologic agents and nursing considerations. Clin J Oncol Nurs. 2018;22(3):350-353. doi:10.1188/18.CJON.350-353

16. Nichols HB, Stürmer T, Lee VS, et al. Breast cancer chemoprevention in an integrated health care setting. JCO Clin Cancer Inform. 2017;1:1-12. doi:10.1200/CCI.16.00059

17. Bevers TB, Helvie M, Bonaccio E, et al. Breast cancer screening and diagnosis, Version 3.2018, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2018;16(11):1362-1389. doi:10.6004/jnccn.2018.0083

18. Visvanathan K, Hurley P, Bantug E, et al. Use of pharmacologic interventions for breast cancer risk reduction: American Society of Clinical Oncology clinical practice guideline [published correction appears in J Clin Oncol. 2013 Dec 1;31(34):4383]. J Clin Oncol. 2013;31(23):2942-2962. doi:10.1200/JCO.2013.49.3122

19. Sealy-Jefferson S, Roseland ME, Cote ML, et al. rural-urban residence and stage at breast cancer diagnosis among postmenopausal women: The Women’s Health Initiative. J Womens Health (Larchmt). 2019;28(2):276-283. doi:10.1089/jwh.2017.6884

20. Holder KA. Veterans in rural America: 2011-2015. Published January 25, 2017. Accessed April 12, 2021. https://www.census.gov/library/publications/2017/acs/acs-36.html

21. Owens WL, Gallagher TJ, Kincheloe MJ, Ruetten VL. Implementation in a large health system of a program to identify women at high risk for breast cancer. J Oncol Pract. 2011;7(2):85-88. doi:10.1200/JOP.2010.000107

2. Pivot X, Viguier J, Touboul C, et al. Breast cancer screening controversy: too much or not enough?. Eur J Cancer Prev. 2015;24 Suppl:S73-S76. doi:10.1097/CEJ.0000000000000145

23. Bidassie B, Kovach A, Vallette MA, et al. Breast Cancer risk assessment and chemoprevention use among veterans affairs primary care providers: a national online survey. Mil Med. 2020;185(3-4):512-518. doi:10.1093/milmed/usz291

24. Brewster AM, Davidson NE, McCaskill-Stevens W. Chemoprevention for breast cancer: overcoming barriers to treatment. Am Soc Clin Oncol Educ Book. 2012;85-90. doi:10.14694/EdBook_AM.2012.32.152

25. Meyskens FL Jr, Curt GA, Brenner DE, et al. Regulatory approval of cancer risk-reducing (chemopreventive) drugs: moving what we have learned into the clinic. Cancer Prev Res (Phila). 2011;4(3):311-323. doi:10.1158/1940-6207.CAPR-09-0014

26. Tice JA, Kerlikowske K. Screening and prevention of breast cancer in primary care. Prim Care. 2009;36(3):533-558. doi:10.1016/j.pop.2009.04.003

27. Vogel VG. Selective estrogen receptor modulators and aromatase inhibitors for breast cancer chemoprevention. Curr Drug Targets. 2011;12(13):1874-1887. doi:10.2174/138945011798184164

28. Vogel VG, Costantino JP, Wickerham DL, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial [published correction appears in JAMA. 2006 Dec 27;296(24):2926] [published correction appears in JAMA. 2007 Sep 5;298(9):973]. JAMA. 2006;295(23):2727-2741. doi:10.1001/jama.295.23.joc60074

29. Pruthi S, Heisey RE, Bevers TB. Chemoprevention for breast cancer. Ann Surg Oncol. 2015;22(10):3230-3235. doi:10.1245/s10434-015-4715-9

30. Cuzick J, Sestak I, Forbes JF, et al. Anastrozole for prevention of breast cancer in high-risk postmenopausal women (IBIS-II): an international, double-blind, randomised placebo-controlled trial [published correction appears in Lancet. 2014 Mar 22;383(9922):1040] [published correction appears in Lancet. 2017 Mar 11;389(10073):1010]. Lancet. 2014;383(9922):1041-1048. doi:10.1016/S0140-6736(13)62292-8

31. Bozovic-Spasojevic I, Azambuja E, McCaskill-Stevens W, Dinh P, Cardoso F. Chemoprevention for breast cancer. Cancer Treat Rev. 2012;38(5):329-339. doi:10.1016/j.ctrv.2011.07.005

32. Gabriel EM, Jatoi I. Breast cancer chemoprevention. Expert Rev Anticancer Ther. 2012;12(2):223-228. doi:10.1586/era.11.206

<--pagebreak-->

33. Crew KD, Albain KS, Hershman DL, Unger JM, Lo SS. How do we increase uptake of tamoxifen and other anti-estrogens for breast cancer prevention?. NPJ Breast Cancer. 2017;3:20. Published 2017 May 19. doi:10.1038/s41523-017-0021-y

34. Ropka ME, Keim J, Philbrick JT. Patient decisions about breast cancer chemoprevention: a systematic review and meta-analysis. J Clin Oncol. 2010;28(18):3090-3095. doi:10.1200/JCO.2009.27.8077

35. Smith SG, Sestak I, Forster A, et al. Factors affecting uptake and adherence to breast cancer chemoprevention: a systematic review and meta-analysis. Ann Oncol. 2016;27(4):575-590. doi:10.1093/annonc/mdv590

36. Grann VR, Patel PR, Jacobson JS, et al. Comparative effectiveness of screening and prevention strategies among BRCA1/2-affected mutation carriers. Breast Cancer Res Treat. 2011 Feb;125(3):837-847. doi:10.1007/s10549-010-1043-4

37. Goss PE, Ingle JN, Alés-Martínez JE, et al. Exemestane for breast-cancer prevention in postmenopausal women [published correction appears in N Engl J Med. 2011 Oct 6;365(14):1361]. N Engl J Med. 2011;364(25):2381-2391. doi:10.1056/NEJMoa1103507

38. Kmietowicz Z. Five in six women reject drugs that could reduce their risk of breast cancer. BMJ. 2015;351:h6650. Published 2015 Dec 8. doi:10.1136/bmj.h6650

39. Nelson HD, Fu R, Griffin JC, Nygren P, Smith ME, Humphrey L. Systematic review: comparative effectiveness of medications to reduce risk for primary breast cancer. Ann Intern Med. 2009;151(10):703-235. doi:10.7326/0003-4819-151-10-200911170-00147

40. Dahabreh IJ, Wieland LS, Adam GP, Halladay C, Lau J, Trikalinos TA. Core needle and open surgery biopsy for diagnosis of breast lesions: an update to the 2009 report. Published September 2014. Accessed April 12, 2021. https://www.ncbi.nlm.nih.gov/books/NBK246878

41. National Cancer Institute. Genetics of breast and ovarian cancer (PDQ)—health profession version. Updated February 12, 2021. Accessed April 12, 2021. http://www.cancer.gov/cancertopics/pdq/genetics/breast-and-ovarian/HealthProfessional

42. US Department of Health and Human Services. National Institutes of Health, National Institute of Environmental Health Sciences The sister study. Accessed April 12, 2021. https://sisterstudy.niehs.nih.gov/english/NIEHS.htm

43. Tutt A, Ashworth A. Can genetic testing guide treatment in breast cancer?. Eur J Cancer. 2008;44(18):2774-2780. doi:10.1016/j.ejca.2008.10.009

44. Katz SJ, Ward KC, Hamilton AS, et al. Gaps in receipt of clinically indicated genetic counseling after diagnosis of breast cancer. J Clin Oncol. 2018;36(12):1218-1224. doi:10.1200/JCO.2017.76.2369

45. US Department of Veterans Affairs. PTSD: National Center for PTSD. How common is PTSD in adults? Updated October 17, 2019. Accessed April 12, 2021. https://www.ptsd.va.gov/understand/common/common_adults.asp

46. US Department of Veterans Affairs. PTSD: National Center for PTSD. How common is PTSD in women? Updated October 16, 2019. Accessed April 12, 2021. https://www.ptsd.va.gov/understand/common/common_women.asp

47. US Department of Veterans Affairs. PTSD: National Center for PTSD. How common is PTSD in veterans? Updated September 24, 2018. Accessed April 12, 2021. https://www.ptsd.va.gov/understand/common/common_veterans.asp

48. Lindberg NM, Wellisch D. Anxiety and compliance among women at high risk for breast cancer. Ann Behav Med. 2001;23(4):298-303. doi:10.1207/S15324796ABM2304_9

49. DiMatteo MR, Lepper HS, Croghan TW. Depression is a risk factor for noncompliance with medical treatment: meta-analysis of the effects of anxiety and depression on patient adherence. Arch Intern Med. 2000;160(14):2101-2107. doi:10.1001/archinte.160.14.2101

50. Centers for Disease Control and Prevention. MMWR appendix: breast cancer rates among black women and white women. Updated October 13, 2016. Accessed April 12, 2021. https://www.cdc.gov/cancer/breast/statistics/trends_invasive.htm

51. Richardson LC, Henley SJ, Miller JW, Massetti G, Thomas CC. Patterns and trends in age-specific black-white differences in breast cancer incidence and mortality - United States, 1999-2014. MMWR Morb Mortal Wkly Rep. 2016;65(40):1093-1098. Published 2016 Oct 14. doi:10.15585/mmwr.mm6540a1

52. Brody JG, Moysich KB, Humblet O, Attfield KR, Beehler GP, Rudel RA. Environmental pollutants and breast cancer: epidemiologic studies. Cancer. 2007;109(12 Suppl):2667-2711. doi:10.1002/cncr.22655

53. Brophy JT, Keith MM, Watterson A, et al. Breast cancer risk in relation to occupations with exposure to carcinogens and endocrine disruptors: a Canadian case-control study. Environ Health. 2012;11:87. Published 2012 Nov 19. doi:10.1186/1476-069X-11-87

54. Labrèche F, Goldberg MS, Valois MF, Nadon L. Postmenopausal breast cancer and occupational exposures. Occup Environ Med. 2010;67(4):263-269. doi:10.1136/oem.2009.049817

55. National Institute of Environmental Health Sciences, Interagency Breast Cancer & Environmental Research Coordinating Committee. Breast cancer and the environment: prioritizing prevention. Updated March 8, 2013. Accessed April 12, 2021. https://www.niehs.nih.gov/about/boards/ibcercc/index.cfm

56. Gail MH, Costantino JP, Pee D, et al. Projecting individualized absolute invasive breast cancer risk in African American women [published correction appears in J Natl Cancer Inst. 2008 Aug 6;100(15):1118] [published correction appears in J Natl Cancer Inst. 2008 Mar 5;100(5):373]. J Natl Cancer Inst. 2007;99(23):1782-1792. doi:10.1093/jnci/djm223

57. Corbie-Smith G, Thomas SB, Williams MV, Moody-Ayers S. Attitudes and beliefs of African Americans toward participation in medical research. J Gen Intern Med. 1999;14(9):537-546. doi:10.1046/j.1525-1497.1999.07048.x

58. Braunstein JB, Sherber NS, Schulman SP, Ding EL, Powe NR. Race, medical researcher distrust, perceived harm, and willingness to participate in cardiovascular prevention trials. Medicine (Baltimore). 2008;87(1):1-9. doi:10.1097/MD.0b013e3181625d78

Article PDF
Author and Disclosure Information

Yeun-Hee Anna Park is Chief of Hematology/Oncology; Alison Keller is a Research Coordinator; and Ta-Chueh Melody Hsu is a Research Nurse Practitioner, all at James J. Peters Veterans Affairs Medical Center, Bronx, New York. Balmatee Bidassie is an Industrial Engineer VA Center for Applied Systems Engineering (VA-CASE), VISN11 - Veterans Engineering Resource Center (VERC) at Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana. Vickie Venne was a Senior Genetic Counselor for the US Department of Veterans Affairs (VA) Genomic Medicine Services, and Sarah Colonna is a Hematologist/Oncologist; both at George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah. Douglas Hawley is a Hematologist/Oncologist at Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio. Lori Hoffman-Högg is a ONS Clinical Nurse Advisor for the Oncology Field Advisory Committee and VHA National Program Manager for Prevention Policy at Veterans Health Administration (VHA) National Center for Health Promotion and Disease Prevention, Durham, North Carolina and VHA Office of Nursing Services, Washington, DC. Bernadette Heron is a Program Manager at Veterans Health Administration, Pharmacy Benefits Management Services in Hines, Illinois. Anita Aggarwal is a Hematologist/Oncologist Washington Veterans Affairs Medical Center, Washington, DC.

Issue
Federal Practitioner - 38(2)s
Publications
Topics
Page Number
S35-S41
Sections
Author and Disclosure Information

Yeun-Hee Anna Park is Chief of Hematology/Oncology; Alison Keller is a Research Coordinator; and Ta-Chueh Melody Hsu is a Research Nurse Practitioner, all at James J. Peters Veterans Affairs Medical Center, Bronx, New York. Balmatee Bidassie is an Industrial Engineer VA Center for Applied Systems Engineering (VA-CASE), VISN11 - Veterans Engineering Resource Center (VERC) at Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana. Vickie Venne was a Senior Genetic Counselor for the US Department of Veterans Affairs (VA) Genomic Medicine Services, and Sarah Colonna is a Hematologist/Oncologist; both at George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah. Douglas Hawley is a Hematologist/Oncologist at Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio. Lori Hoffman-Högg is a ONS Clinical Nurse Advisor for the Oncology Field Advisory Committee and VHA National Program Manager for Prevention Policy at Veterans Health Administration (VHA) National Center for Health Promotion and Disease Prevention, Durham, North Carolina and VHA Office of Nursing Services, Washington, DC. Bernadette Heron is a Program Manager at Veterans Health Administration, Pharmacy Benefits Management Services in Hines, Illinois. Anita Aggarwal is a Hematologist/Oncologist Washington Veterans Affairs Medical Center, Washington, DC.

Author and Disclosure Information

Yeun-Hee Anna Park is Chief of Hematology/Oncology; Alison Keller is a Research Coordinator; and Ta-Chueh Melody Hsu is a Research Nurse Practitioner, all at James J. Peters Veterans Affairs Medical Center, Bronx, New York. Balmatee Bidassie is an Industrial Engineer VA Center for Applied Systems Engineering (VA-CASE), VISN11 - Veterans Engineering Resource Center (VERC) at Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana. Vickie Venne was a Senior Genetic Counselor for the US Department of Veterans Affairs (VA) Genomic Medicine Services, and Sarah Colonna is a Hematologist/Oncologist; both at George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah. Douglas Hawley is a Hematologist/Oncologist at Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio. Lori Hoffman-Högg is a ONS Clinical Nurse Advisor for the Oncology Field Advisory Committee and VHA National Program Manager for Prevention Policy at Veterans Health Administration (VHA) National Center for Health Promotion and Disease Prevention, Durham, North Carolina and VHA Office of Nursing Services, Washington, DC. Bernadette Heron is a Program Manager at Veterans Health Administration, Pharmacy Benefits Management Services in Hines, Illinois. Anita Aggarwal is a Hematologist/Oncologist Washington Veterans Affairs Medical Center, Washington, DC.

Article PDF
Article PDF

The number of women seeking care from the Veterans Health Administration (VHA) is increasing.1 In 2015, there were 2 million women veterans in the United States, which is 9.4% of the total veteran population. This group is expected to increase at an average of about 18,000 women per year for the next 10 years.2 The percentage of women veterans who are US Department of Veterans Affairs (VA) users aged 45 to 64 years rose 46% from 2000 to 2015.1,3-4 It is estimated that 15% of veterans who used VA services in 2020 were women.1 Nineteen percent of women veterans are Black.1 The median age of women veterans in 2015 was 50 years.5 Breast cancer is the leading cancer affecting female veterans, and data suggest they have an increased risk of breast cancer based on unique service-related exposures.1,6-9

In the US, about 10 million women are eligible for breast cancer preventive therapy, including, but not limited to, medications, surgery, or lifestyle changes.10 Secondary prevention options include change in surveillance that can reduce their risk or identify cancer at an earlier stage when treatment is more effective. The United States Preventive Services Task Force, the National Comprehensive Cancer Network, the American Society for Clinical Oncology, the National Institute for Health and Care Excellence, and the Oncology Nursing Society recommend screening women aged ≥ 35 years to assess breast cancer risk.11-18 If a woman is at increased risk, she may be a candidate for chemoprevention, prozphylactic surgery, and possibly an enhanced screening regimen.

Urban and minority women are an understudied population. Most veterans (75%) live in urban or suburban settings.19,20 Urban veteran women constitute an important potential study population.

Chemoprevention measures have been underused because of factors involving both women and their health care providers. A large proportion of women are unaware of their higher risk status due to lack of adequate screening and risk assessment.21,22 In addition to patient lack of awareness of their high-risk status, primary care physicians are also reluctant to prescribe chemopreventive agents due to a lack of comfort or familiarity with the risks and benefits.23-26 The STAR2015, BCPT2005, IBIS2014, MAP3 2011, IBIS-I 2014, and IBIS II 2014 studies clearly demonstrate a 49 to 62% reduction in risk for women using chemoprevention such as selective estrogen receptor modulators or aromatase inhibitors, respectively.27-32 Yet only 4 to 9% of high-risk women not enrolled in a clinical trial are using chemoprevention.33-39

The possibility of developing breast cancer also may be increased because of a positive family history or being a member of a family in which there is a known susceptibility gene mutation.40 Based on these risk factors, women may be eligible for tailored follow-up and genetic counseling.41-44

Nationally, 7 to 10% of the civilian US population will experience posttraumatic stress disorder (PTSD).45 The rates are remarkably higher for women veterans, with roughly 20% diagnosed with PTSD.46,47 Anxiety and PTSD have been implicated in poor adherence to medical advice.48,49

In 2014, a national VA multidisciplinary group focused on breast cancer prevention, detection, treatment, and research to address breast health in the growing population of women veterans. High-risk breast cancer screenings are not routinely carried out by the VA in primary care, women’s health, or oncology services. Furthermore, the recording of screening questionnaire results was not synchronized until a standard questionnaire was created and approved as a template by this group in the VA electronic medical record (EMR) in 2015.

Several prediction models can identify which women are at an increased risk of developing breast cancer. The most commonly used risk assessment model, the Gail breast cancer risk assessment tool (BCRAT), has been refined to include women of additional ethnicities (https://www.cancer.gov/bcrisktool).

This pilot project was launched to identify an effective manner to screen women veterans regarding their risk of developing breast cancer and refer them for chemoprevention education or genetic counseling as appropriate.

 

 

Methods

A high-risk breast cancer screening questionnaire based on the Gail BCRAT and including lifestyle questions was developed and included as a note template in the VA EMR. The James J. Peters VA Medical Center, Bronx, NY (JJPVAMC) and the Washington DC VA Medical Center (DCVAMC) ran a pilot study between 2015 and 2018 using this breast cancer screening questionnaire to collect data from women veterans. Quality Executive Committee and institutional review board approvals were granted respectively.

Eligibility criteria included women aged ≥ 35 years with no personal history of breast cancer. Most patients were self-referred, but participants also were recruited during VA Breast Cancer Awareness month events, health fairs, or at informational tables in the hospital lobbies. After completing the 20 multiple choice questionnaire with a study team member, either in person or over the phone, a 5-year and lifetime risk of invasive breast cancer was calculated using the Gail BCRAT. A woman is considered high risk and eligible for chemoprevention if her 5-year risk is > 1.66% or her lifetime risk is ≥ 20%. Eligibility for genetic counseling is based on the Breast Cancer Referral Screening Tool, which includes a personal or family history of breast or ovarian cancer and Jewish ancestry.

All patients were notified of their average or high risk status by a clinician. Those who were deemed to be average risk received a follow-up letter in the mail with instructions (eg, to follow-up with a yearly mammogram). Those who were deemed to be high risk for developing breast cancer were asked to come in for an appointment with the study principal investigator (a VA oncologist/breast cancer specialist) to discuss prevention options, further screening, or referrals to genetic counseling. Depending on a patient’s other health factors, a woman at high risk for developing breast cancer also may be a candidate for chemoprevention with tamoxifen, raloxifene, exemestane, anastrozole, or letrozole.

Data on the participant’s lifestyle, including exercise, diet, and smoking, were evaluated to determine whether these factors had an impact on risk status.

Results

The JJP and DC VAMCs screened 103 women veterans between 2015 and 2018. Four patients were excluded for nonveteran (spousal) status, leaving 99 women veterans with a mean age of 54 years. The most common self-reported races were Black (60%), non-Hispanic White (14%), and Hispanic or Latino (13%) (Table 1).

Women veterans in our study were nearly 3-times more likely than the general population were to receive a high-risk Gail Score/BCRAT (35% vs 13%, respectively).50,51 Of this subset, 46% had breast biopsies, and 86% had a positive family history. Thirty-one percent of Black women in our study were high risk, while nationally, 8.2 to 13.3% of Black women aged 50 to 59 years are considered high risk.50,51 Of the Black high-risk group with a high Gail/BCRAT score, 94% had a positive family history, and 33% had a history of breast biopsy (Table 2).

Of the 35 high-risk patients 26 (74%) patients accepted consultations for chemoprevention and 5 (19%) started chemoprevention. Of this high-risk group, 13 (37%) patients were referred for genetic counseling (Table 3).44 The prevalence of PTSD was present in 31% of high-risk women and 29% of the cohort (Figure).The lifestyle questions indicated that, among all participants, 79% had an overweight or obese body mass index; 58% exercised weekly; 51% consumed alcohol; 14% were smokers; and 21% consumed 3 to 4 servings of fruits/vegetables daily.

 

 

Discussion

Breast cancer is the most common cancer in women.52 The number of women with breast cancer in the VHA has more than tripled from 1995 to 2012.1 The lifetime risk of developing breast cancer in the general population is about 13%.50 This rate can be affected by risk factors including age, hormone exposure, family history, radiation exposure, and lifestyle factors, such as weight and alcohol use.6,52-56 In the United States, invasive breast cancer affects 1 in 8 women.50,52,57

Our screened population showed nearly 3 times as many women veterans were at an increased risk for breast cancer when compared with historical averages in US women. This difference may be based on a high rate of prior breast biopsies or positive family history, although a provocative study using the Surveillance, Epidemiology, and End Results database showed military women to have higher rates of breast cancer as well.9 Historically, Blacks are vastly understudied in clinical research with only 5% representation on a national level.5,58 The urban locations of both pilot sites (Washington, DC and Bronx, NY) allowed for the inclusion of minority patients in our study. We found that the rates of breast cancer in Black women veterans to be higher than seen nationally, possibly prompting further screening initiatives for this understudied population.

Our pilot study’s chemoprevention utilization (19%) was double the < 10% seen in the national population.33-35 The presence of a knowledgeable breast health practitioner to recruit study participants and offer personalized counseling to women veterans is a likely factor in overcoming barriers to chemopreventive acceptance. These participants may have been motivated to seek care for their high-risk status given a strong family history and prior breast biopsies.

Interestingly, a 3-fold higher PTSD rate was seen in this pilot population (29%) when compared with PTSD rates in the general female population (7-10%) and still one-third higher than the general population of women veterans (20%).45-47 Mental health, anxiety, and PTSD have been barriers to patients who sought treatment and have been implicated in poor adherence to medical advice.48,49 Cancer screening can induce anxiety in patients, and it may be amplified in patients with PTSD. It was remarkable that although adherence with screening recommendations is decreased when PTSD is present, our patient population demonstrated a higher rate of screening adherence.

Women who are seen at the VA often use multiple clinical specialties, and their EMR can be accessed across VA medical centers nationwide. Therefore, identifying women veterans who meet screening criteria is easily attainable within the VA.

When comparing high-risk with average risk women, the lifestyle results (BMI, smoking history, exercise and consumption of fruits, vegetables and alcohol) were essentially the same. Lifestyle factors were similar to national population rates and were unlikely to impact risk levels.

Limitations

Study limitations included a high number of self-referrals and the large percentage of patients with a family history of breast cancer, making them more likely to seek screening. The higher-than-average risk of breast cancer may be driven by a high rate of breast biopsies and a strong family history. Lifestyle metrics could not be accurately compared to other national assessments of lifestyle factors due to the difference in data points that we used or the format of our questions.

 

 

Conclusions

As the number of women veterans increases and the incidence of breast cancer in women veterans rise, chemoprevention options should follow national guidelines. To our knowledge, this is the only oncology study with 60% Black women veterans. This study had a higher participation rate for Black women veterans than is typically seen in national research studies and shows the VA to be a germane source for further understanding of an understudied population that may benefit from increased screening for breast cancer.

A team-based, multidisciplinary model that meets the unique healthcare needs of women veterans results in a patient-centric delivery of care for assessing breast cancer risk status and prevention options. This model can be replicated nationally by directing primary care physicians and women’s health practitioners to a risk-assessment questionnaire and referring high-risk women for appropriate preventative care. Given that these results show chemoprevention adherence rates doubled those seen nationally, perhaps techniques used within this VA pilot study may be adapted to decrease breast cancer incidence nationally.

Since the rate of PTSD among women veterans is triple the national average, we would expect adherence rates to be lower in our patient cohort. However, the multidisciplinary approach we used in this study (eg, 1:1 consultation with oncologist; genetic counseling referrals; mental health support available), may have improved adherence rates. Perhaps the high rates of PTSD seen in the VA patient population can be a useful way to explore patient adherence rates in those with mental illness and medical conditions.

Future research with a larger cohort may lead to greater insight into the correlation between PTSD and adherence to treatment. Exploring the connection between breast cancer, epigenetics, and specific military service-related exposures could be an area of analysis among this veteran population exhibiting increased breast cancer rates. VAMCs are situated in rural, suburban, and urban locations across the United States and offers a diverse socioeconomic and ethnic patient population for inclusion in clinical investigations. Women veterans make up a small subpopulation of women in the United States, but it is worth considering VA patients as an untapped resource for research collaboration.

Acknowledgements

The authors thank Steven Sanchez and Marissa Vallette, PhD, Breast Health Research Group. This research project was approved by the James J. Peters VA Medical Center Quality Executive Committee and the Washington, DC VA Medical Center Institutional Review Board. This work was supported by the US Department of Veterans Affairs. This work did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author disclosures

The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

The number of women seeking care from the Veterans Health Administration (VHA) is increasing.1 In 2015, there were 2 million women veterans in the United States, which is 9.4% of the total veteran population. This group is expected to increase at an average of about 18,000 women per year for the next 10 years.2 The percentage of women veterans who are US Department of Veterans Affairs (VA) users aged 45 to 64 years rose 46% from 2000 to 2015.1,3-4 It is estimated that 15% of veterans who used VA services in 2020 were women.1 Nineteen percent of women veterans are Black.1 The median age of women veterans in 2015 was 50 years.5 Breast cancer is the leading cancer affecting female veterans, and data suggest they have an increased risk of breast cancer based on unique service-related exposures.1,6-9

In the US, about 10 million women are eligible for breast cancer preventive therapy, including, but not limited to, medications, surgery, or lifestyle changes.10 Secondary prevention options include change in surveillance that can reduce their risk or identify cancer at an earlier stage when treatment is more effective. The United States Preventive Services Task Force, the National Comprehensive Cancer Network, the American Society for Clinical Oncology, the National Institute for Health and Care Excellence, and the Oncology Nursing Society recommend screening women aged ≥ 35 years to assess breast cancer risk.11-18 If a woman is at increased risk, she may be a candidate for chemoprevention, prozphylactic surgery, and possibly an enhanced screening regimen.

Urban and minority women are an understudied population. Most veterans (75%) live in urban or suburban settings.19,20 Urban veteran women constitute an important potential study population.

Chemoprevention measures have been underused because of factors involving both women and their health care providers. A large proportion of women are unaware of their higher risk status due to lack of adequate screening and risk assessment.21,22 In addition to patient lack of awareness of their high-risk status, primary care physicians are also reluctant to prescribe chemopreventive agents due to a lack of comfort or familiarity with the risks and benefits.23-26 The STAR2015, BCPT2005, IBIS2014, MAP3 2011, IBIS-I 2014, and IBIS II 2014 studies clearly demonstrate a 49 to 62% reduction in risk for women using chemoprevention such as selective estrogen receptor modulators or aromatase inhibitors, respectively.27-32 Yet only 4 to 9% of high-risk women not enrolled in a clinical trial are using chemoprevention.33-39

The possibility of developing breast cancer also may be increased because of a positive family history or being a member of a family in which there is a known susceptibility gene mutation.40 Based on these risk factors, women may be eligible for tailored follow-up and genetic counseling.41-44

Nationally, 7 to 10% of the civilian US population will experience posttraumatic stress disorder (PTSD).45 The rates are remarkably higher for women veterans, with roughly 20% diagnosed with PTSD.46,47 Anxiety and PTSD have been implicated in poor adherence to medical advice.48,49

In 2014, a national VA multidisciplinary group focused on breast cancer prevention, detection, treatment, and research to address breast health in the growing population of women veterans. High-risk breast cancer screenings are not routinely carried out by the VA in primary care, women’s health, or oncology services. Furthermore, the recording of screening questionnaire results was not synchronized until a standard questionnaire was created and approved as a template by this group in the VA electronic medical record (EMR) in 2015.

Several prediction models can identify which women are at an increased risk of developing breast cancer. The most commonly used risk assessment model, the Gail breast cancer risk assessment tool (BCRAT), has been refined to include women of additional ethnicities (https://www.cancer.gov/bcrisktool).

This pilot project was launched to identify an effective manner to screen women veterans regarding their risk of developing breast cancer and refer them for chemoprevention education or genetic counseling as appropriate.

 

 

Methods

A high-risk breast cancer screening questionnaire based on the Gail BCRAT and including lifestyle questions was developed and included as a note template in the VA EMR. The James J. Peters VA Medical Center, Bronx, NY (JJPVAMC) and the Washington DC VA Medical Center (DCVAMC) ran a pilot study between 2015 and 2018 using this breast cancer screening questionnaire to collect data from women veterans. Quality Executive Committee and institutional review board approvals were granted respectively.

Eligibility criteria included women aged ≥ 35 years with no personal history of breast cancer. Most patients were self-referred, but participants also were recruited during VA Breast Cancer Awareness month events, health fairs, or at informational tables in the hospital lobbies. After completing the 20 multiple choice questionnaire with a study team member, either in person or over the phone, a 5-year and lifetime risk of invasive breast cancer was calculated using the Gail BCRAT. A woman is considered high risk and eligible for chemoprevention if her 5-year risk is > 1.66% or her lifetime risk is ≥ 20%. Eligibility for genetic counseling is based on the Breast Cancer Referral Screening Tool, which includes a personal or family history of breast or ovarian cancer and Jewish ancestry.

All patients were notified of their average or high risk status by a clinician. Those who were deemed to be average risk received a follow-up letter in the mail with instructions (eg, to follow-up with a yearly mammogram). Those who were deemed to be high risk for developing breast cancer were asked to come in for an appointment with the study principal investigator (a VA oncologist/breast cancer specialist) to discuss prevention options, further screening, or referrals to genetic counseling. Depending on a patient’s other health factors, a woman at high risk for developing breast cancer also may be a candidate for chemoprevention with tamoxifen, raloxifene, exemestane, anastrozole, or letrozole.

Data on the participant’s lifestyle, including exercise, diet, and smoking, were evaluated to determine whether these factors had an impact on risk status.

Results

The JJP and DC VAMCs screened 103 women veterans between 2015 and 2018. Four patients were excluded for nonveteran (spousal) status, leaving 99 women veterans with a mean age of 54 years. The most common self-reported races were Black (60%), non-Hispanic White (14%), and Hispanic or Latino (13%) (Table 1).

Women veterans in our study were nearly 3-times more likely than the general population were to receive a high-risk Gail Score/BCRAT (35% vs 13%, respectively).50,51 Of this subset, 46% had breast biopsies, and 86% had a positive family history. Thirty-one percent of Black women in our study were high risk, while nationally, 8.2 to 13.3% of Black women aged 50 to 59 years are considered high risk.50,51 Of the Black high-risk group with a high Gail/BCRAT score, 94% had a positive family history, and 33% had a history of breast biopsy (Table 2).

Of the 35 high-risk patients 26 (74%) patients accepted consultations for chemoprevention and 5 (19%) started chemoprevention. Of this high-risk group, 13 (37%) patients were referred for genetic counseling (Table 3).44 The prevalence of PTSD was present in 31% of high-risk women and 29% of the cohort (Figure).The lifestyle questions indicated that, among all participants, 79% had an overweight or obese body mass index; 58% exercised weekly; 51% consumed alcohol; 14% were smokers; and 21% consumed 3 to 4 servings of fruits/vegetables daily.

 

 

Discussion

Breast cancer is the most common cancer in women.52 The number of women with breast cancer in the VHA has more than tripled from 1995 to 2012.1 The lifetime risk of developing breast cancer in the general population is about 13%.50 This rate can be affected by risk factors including age, hormone exposure, family history, radiation exposure, and lifestyle factors, such as weight and alcohol use.6,52-56 In the United States, invasive breast cancer affects 1 in 8 women.50,52,57

Our screened population showed nearly 3 times as many women veterans were at an increased risk for breast cancer when compared with historical averages in US women. This difference may be based on a high rate of prior breast biopsies or positive family history, although a provocative study using the Surveillance, Epidemiology, and End Results database showed military women to have higher rates of breast cancer as well.9 Historically, Blacks are vastly understudied in clinical research with only 5% representation on a national level.5,58 The urban locations of both pilot sites (Washington, DC and Bronx, NY) allowed for the inclusion of minority patients in our study. We found that the rates of breast cancer in Black women veterans to be higher than seen nationally, possibly prompting further screening initiatives for this understudied population.

Our pilot study’s chemoprevention utilization (19%) was double the < 10% seen in the national population.33-35 The presence of a knowledgeable breast health practitioner to recruit study participants and offer personalized counseling to women veterans is a likely factor in overcoming barriers to chemopreventive acceptance. These participants may have been motivated to seek care for their high-risk status given a strong family history and prior breast biopsies.

Interestingly, a 3-fold higher PTSD rate was seen in this pilot population (29%) when compared with PTSD rates in the general female population (7-10%) and still one-third higher than the general population of women veterans (20%).45-47 Mental health, anxiety, and PTSD have been barriers to patients who sought treatment and have been implicated in poor adherence to medical advice.48,49 Cancer screening can induce anxiety in patients, and it may be amplified in patients with PTSD. It was remarkable that although adherence with screening recommendations is decreased when PTSD is present, our patient population demonstrated a higher rate of screening adherence.

Women who are seen at the VA often use multiple clinical specialties, and their EMR can be accessed across VA medical centers nationwide. Therefore, identifying women veterans who meet screening criteria is easily attainable within the VA.

When comparing high-risk with average risk women, the lifestyle results (BMI, smoking history, exercise and consumption of fruits, vegetables and alcohol) were essentially the same. Lifestyle factors were similar to national population rates and were unlikely to impact risk levels.

Limitations

Study limitations included a high number of self-referrals and the large percentage of patients with a family history of breast cancer, making them more likely to seek screening. The higher-than-average risk of breast cancer may be driven by a high rate of breast biopsies and a strong family history. Lifestyle metrics could not be accurately compared to other national assessments of lifestyle factors due to the difference in data points that we used or the format of our questions.

 

 

Conclusions

As the number of women veterans increases and the incidence of breast cancer in women veterans rise, chemoprevention options should follow national guidelines. To our knowledge, this is the only oncology study with 60% Black women veterans. This study had a higher participation rate for Black women veterans than is typically seen in national research studies and shows the VA to be a germane source for further understanding of an understudied population that may benefit from increased screening for breast cancer.

A team-based, multidisciplinary model that meets the unique healthcare needs of women veterans results in a patient-centric delivery of care for assessing breast cancer risk status and prevention options. This model can be replicated nationally by directing primary care physicians and women’s health practitioners to a risk-assessment questionnaire and referring high-risk women for appropriate preventative care. Given that these results show chemoprevention adherence rates doubled those seen nationally, perhaps techniques used within this VA pilot study may be adapted to decrease breast cancer incidence nationally.

Since the rate of PTSD among women veterans is triple the national average, we would expect adherence rates to be lower in our patient cohort. However, the multidisciplinary approach we used in this study (eg, 1:1 consultation with oncologist; genetic counseling referrals; mental health support available), may have improved adherence rates. Perhaps the high rates of PTSD seen in the VA patient population can be a useful way to explore patient adherence rates in those with mental illness and medical conditions.

Future research with a larger cohort may lead to greater insight into the correlation between PTSD and adherence to treatment. Exploring the connection between breast cancer, epigenetics, and specific military service-related exposures could be an area of analysis among this veteran population exhibiting increased breast cancer rates. VAMCs are situated in rural, suburban, and urban locations across the United States and offers a diverse socioeconomic and ethnic patient population for inclusion in clinical investigations. Women veterans make up a small subpopulation of women in the United States, but it is worth considering VA patients as an untapped resource for research collaboration.

Acknowledgements

The authors thank Steven Sanchez and Marissa Vallette, PhD, Breast Health Research Group. This research project was approved by the James J. Peters VA Medical Center Quality Executive Committee and the Washington, DC VA Medical Center Institutional Review Board. This work was supported by the US Department of Veterans Affairs. This work did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author disclosures

The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

References

1. US Department of Veterans Affairs. National Center for Veterans Analysis and Statistics. The past, present and future of women veterans. Published February 2017. Accessed April 28, 2021. https://www.va.gov/vetdata/docs/specialreports/women_veterans_2015_final.pdf.

2. Frayne SM, Carney DV, Bastian L, et al. The VA Women’s Health Practice-Based Research Network: amplifying women veterans’ voices in VA research. J Gen Intern Med. 2013;28 Suppl 2(Suppl 2):S504-S509. doi:10.1007/s11606-013-2476-3

3. US Department of Veterans Affairs, Veterans Health Administration, Women’s Health Evaluation Initiative, Women Veterans Health Strategic Health Care Group. Sourcebook: women veterans in the Veterans Health Administration. Volume 1: Sociodemographic characteristics and use of VHA care. Published December 2010. Accessed April 12, 2021. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=2455

4. Bean-Mayberry B, Yano EM, Bayliss N, Navratil J, Weisman CS, Scholle SH. Federally funded comprehensive women’s health centers: leading innovation in women’s healthcare delivery. J Womens Health (Larchmt). 2007;16(9):1281-1290. doi:10.1089/jwh.2006.0284

5. US Department of Veterans Affairs. National Center for Veterans Analysis and Statistics.VA utilization profile FY 2016. Published November 2017. Accessed April 12, 2021. https://www.va.gov/vetdata/docs/QuickFacts/VA_Utilization_Profile.PDF

6. Ekenga CC, Parks CG, Sandler DP. Chemical exposures in the workplace and breast cancer risk: a prospective cohort study. Int J Cancer. 2015;137(7):1765-1774. doi:10.1002/ijc.29545

7. Rennix CP, Quinn MM, Amoroso PJ, Eisen EA, Wegman DH. Risk of breast cancer among enlisted Army women occupationally exposed to volatile organic compounds. Am J Ind Med. 2005;48(3):157-167. doi:10.1002/ajim.20201

8. Ritz B. Cancer mortality among workers exposed to chemicals during uranium processing. J Occup Environ Med. 1999;41(7):556-566. doi:10.1097/00043764-199907000-00004

9. Zhu K, Devesa SS, Wu H, et al. Cancer incidence in the U.S. military population: comparison with rates from the SEER program. Cancer Epidemiol Biomarkers Prev. 2009;18(6):1740-1745. doi:10.1158/1055-9965.EPI-09-0041

10. Freedman AN, Yu B, Gail MH, et al. Benefit/risk assessment for breast cancer chemoprevention with raloxifene or tamoxifen for women age 50 years or older [published correction appears in J Clin Oncol. 2013 Nov 10;31(32):4167]. J Clin Oncol. 2011;29(17):2327-2333. doi:10.1200/JCO.2010.33.0258

11. Greene, H. Cancer prevention, screening and early detection. In: Gobel BH, Triest-Robertson S, Vogel WH, eds. Advanced Oncology Nursing Certification Review and Resource Manual. 3rd ed. Oncology Nursing Society; 2016:1-34. https://www.ons.org/sites/default/files/publication_pdfs/2%20ADVPrac%20chapter%201.pdf

12. National Comprehensive Cancer Network. NCCN Breast Cancer Risk Reduction. Version 1.2021 NCCN Clinical Practice Guidelines in Oncology. Updated March 24, 2021 Accessed April 12, 2021. https://www.nccn.org/professionals/physician_gls/pdf/breast_risk.pdf

13. US Preventive Services Task Force. Breast cancer: Medications use to reduce risk. Updated September 3, 2019. Accessed April 12, 2021. https://www.uspreventiveservicestaskforce.org/uspstf/recommendation/breast-cancer-medications-for-risk-reduction

14. Moyer VA; U.S. Preventive Services Task Force. Medications to decrease the risk for breast cancer in women: recommendations from the U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2013;159(10):698-708. doi:10.7326/0003-4819-159-10-201311190-00717

15. Boucher JE. Chemoprevention: an overview of pharmacologic agents and nursing considerations. Clin J Oncol Nurs. 2018;22(3):350-353. doi:10.1188/18.CJON.350-353

16. Nichols HB, Stürmer T, Lee VS, et al. Breast cancer chemoprevention in an integrated health care setting. JCO Clin Cancer Inform. 2017;1:1-12. doi:10.1200/CCI.16.00059

17. Bevers TB, Helvie M, Bonaccio E, et al. Breast cancer screening and diagnosis, Version 3.2018, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2018;16(11):1362-1389. doi:10.6004/jnccn.2018.0083

18. Visvanathan K, Hurley P, Bantug E, et al. Use of pharmacologic interventions for breast cancer risk reduction: American Society of Clinical Oncology clinical practice guideline [published correction appears in J Clin Oncol. 2013 Dec 1;31(34):4383]. J Clin Oncol. 2013;31(23):2942-2962. doi:10.1200/JCO.2013.49.3122

19. Sealy-Jefferson S, Roseland ME, Cote ML, et al. rural-urban residence and stage at breast cancer diagnosis among postmenopausal women: The Women’s Health Initiative. J Womens Health (Larchmt). 2019;28(2):276-283. doi:10.1089/jwh.2017.6884

20. Holder KA. Veterans in rural America: 2011-2015. Published January 25, 2017. Accessed April 12, 2021. https://www.census.gov/library/publications/2017/acs/acs-36.html

21. Owens WL, Gallagher TJ, Kincheloe MJ, Ruetten VL. Implementation in a large health system of a program to identify women at high risk for breast cancer. J Oncol Pract. 2011;7(2):85-88. doi:10.1200/JOP.2010.000107

2. Pivot X, Viguier J, Touboul C, et al. Breast cancer screening controversy: too much or not enough?. Eur J Cancer Prev. 2015;24 Suppl:S73-S76. doi:10.1097/CEJ.0000000000000145

23. Bidassie B, Kovach A, Vallette MA, et al. Breast Cancer risk assessment and chemoprevention use among veterans affairs primary care providers: a national online survey. Mil Med. 2020;185(3-4):512-518. doi:10.1093/milmed/usz291

24. Brewster AM, Davidson NE, McCaskill-Stevens W. Chemoprevention for breast cancer: overcoming barriers to treatment. Am Soc Clin Oncol Educ Book. 2012;85-90. doi:10.14694/EdBook_AM.2012.32.152

25. Meyskens FL Jr, Curt GA, Brenner DE, et al. Regulatory approval of cancer risk-reducing (chemopreventive) drugs: moving what we have learned into the clinic. Cancer Prev Res (Phila). 2011;4(3):311-323. doi:10.1158/1940-6207.CAPR-09-0014

26. Tice JA, Kerlikowske K. Screening and prevention of breast cancer in primary care. Prim Care. 2009;36(3):533-558. doi:10.1016/j.pop.2009.04.003

27. Vogel VG. Selective estrogen receptor modulators and aromatase inhibitors for breast cancer chemoprevention. Curr Drug Targets. 2011;12(13):1874-1887. doi:10.2174/138945011798184164

28. Vogel VG, Costantino JP, Wickerham DL, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial [published correction appears in JAMA. 2006 Dec 27;296(24):2926] [published correction appears in JAMA. 2007 Sep 5;298(9):973]. JAMA. 2006;295(23):2727-2741. doi:10.1001/jama.295.23.joc60074

29. Pruthi S, Heisey RE, Bevers TB. Chemoprevention for breast cancer. Ann Surg Oncol. 2015;22(10):3230-3235. doi:10.1245/s10434-015-4715-9

30. Cuzick J, Sestak I, Forbes JF, et al. Anastrozole for prevention of breast cancer in high-risk postmenopausal women (IBIS-II): an international, double-blind, randomised placebo-controlled trial [published correction appears in Lancet. 2014 Mar 22;383(9922):1040] [published correction appears in Lancet. 2017 Mar 11;389(10073):1010]. Lancet. 2014;383(9922):1041-1048. doi:10.1016/S0140-6736(13)62292-8

31. Bozovic-Spasojevic I, Azambuja E, McCaskill-Stevens W, Dinh P, Cardoso F. Chemoprevention for breast cancer. Cancer Treat Rev. 2012;38(5):329-339. doi:10.1016/j.ctrv.2011.07.005

32. Gabriel EM, Jatoi I. Breast cancer chemoprevention. Expert Rev Anticancer Ther. 2012;12(2):223-228. doi:10.1586/era.11.206

<--pagebreak-->

33. Crew KD, Albain KS, Hershman DL, Unger JM, Lo SS. How do we increase uptake of tamoxifen and other anti-estrogens for breast cancer prevention?. NPJ Breast Cancer. 2017;3:20. Published 2017 May 19. doi:10.1038/s41523-017-0021-y

34. Ropka ME, Keim J, Philbrick JT. Patient decisions about breast cancer chemoprevention: a systematic review and meta-analysis. J Clin Oncol. 2010;28(18):3090-3095. doi:10.1200/JCO.2009.27.8077

35. Smith SG, Sestak I, Forster A, et al. Factors affecting uptake and adherence to breast cancer chemoprevention: a systematic review and meta-analysis. Ann Oncol. 2016;27(4):575-590. doi:10.1093/annonc/mdv590

36. Grann VR, Patel PR, Jacobson JS, et al. Comparative effectiveness of screening and prevention strategies among BRCA1/2-affected mutation carriers. Breast Cancer Res Treat. 2011 Feb;125(3):837-847. doi:10.1007/s10549-010-1043-4

37. Goss PE, Ingle JN, Alés-Martínez JE, et al. Exemestane for breast-cancer prevention in postmenopausal women [published correction appears in N Engl J Med. 2011 Oct 6;365(14):1361]. N Engl J Med. 2011;364(25):2381-2391. doi:10.1056/NEJMoa1103507

38. Kmietowicz Z. Five in six women reject drugs that could reduce their risk of breast cancer. BMJ. 2015;351:h6650. Published 2015 Dec 8. doi:10.1136/bmj.h6650

39. Nelson HD, Fu R, Griffin JC, Nygren P, Smith ME, Humphrey L. Systematic review: comparative effectiveness of medications to reduce risk for primary breast cancer. Ann Intern Med. 2009;151(10):703-235. doi:10.7326/0003-4819-151-10-200911170-00147

40. Dahabreh IJ, Wieland LS, Adam GP, Halladay C, Lau J, Trikalinos TA. Core needle and open surgery biopsy for diagnosis of breast lesions: an update to the 2009 report. Published September 2014. Accessed April 12, 2021. https://www.ncbi.nlm.nih.gov/books/NBK246878

41. National Cancer Institute. Genetics of breast and ovarian cancer (PDQ)—health profession version. Updated February 12, 2021. Accessed April 12, 2021. http://www.cancer.gov/cancertopics/pdq/genetics/breast-and-ovarian/HealthProfessional

42. US Department of Health and Human Services. National Institutes of Health, National Institute of Environmental Health Sciences The sister study. Accessed April 12, 2021. https://sisterstudy.niehs.nih.gov/english/NIEHS.htm

43. Tutt A, Ashworth A. Can genetic testing guide treatment in breast cancer?. Eur J Cancer. 2008;44(18):2774-2780. doi:10.1016/j.ejca.2008.10.009

44. Katz SJ, Ward KC, Hamilton AS, et al. Gaps in receipt of clinically indicated genetic counseling after diagnosis of breast cancer. J Clin Oncol. 2018;36(12):1218-1224. doi:10.1200/JCO.2017.76.2369

45. US Department of Veterans Affairs. PTSD: National Center for PTSD. How common is PTSD in adults? Updated October 17, 2019. Accessed April 12, 2021. https://www.ptsd.va.gov/understand/common/common_adults.asp

46. US Department of Veterans Affairs. PTSD: National Center for PTSD. How common is PTSD in women? Updated October 16, 2019. Accessed April 12, 2021. https://www.ptsd.va.gov/understand/common/common_women.asp

47. US Department of Veterans Affairs. PTSD: National Center for PTSD. How common is PTSD in veterans? Updated September 24, 2018. Accessed April 12, 2021. https://www.ptsd.va.gov/understand/common/common_veterans.asp

48. Lindberg NM, Wellisch D. Anxiety and compliance among women at high risk for breast cancer. Ann Behav Med. 2001;23(4):298-303. doi:10.1207/S15324796ABM2304_9

49. DiMatteo MR, Lepper HS, Croghan TW. Depression is a risk factor for noncompliance with medical treatment: meta-analysis of the effects of anxiety and depression on patient adherence. Arch Intern Med. 2000;160(14):2101-2107. doi:10.1001/archinte.160.14.2101

50. Centers for Disease Control and Prevention. MMWR appendix: breast cancer rates among black women and white women. Updated October 13, 2016. Accessed April 12, 2021. https://www.cdc.gov/cancer/breast/statistics/trends_invasive.htm

51. Richardson LC, Henley SJ, Miller JW, Massetti G, Thomas CC. Patterns and trends in age-specific black-white differences in breast cancer incidence and mortality - United States, 1999-2014. MMWR Morb Mortal Wkly Rep. 2016;65(40):1093-1098. Published 2016 Oct 14. doi:10.15585/mmwr.mm6540a1

52. Brody JG, Moysich KB, Humblet O, Attfield KR, Beehler GP, Rudel RA. Environmental pollutants and breast cancer: epidemiologic studies. Cancer. 2007;109(12 Suppl):2667-2711. doi:10.1002/cncr.22655

53. Brophy JT, Keith MM, Watterson A, et al. Breast cancer risk in relation to occupations with exposure to carcinogens and endocrine disruptors: a Canadian case-control study. Environ Health. 2012;11:87. Published 2012 Nov 19. doi:10.1186/1476-069X-11-87

54. Labrèche F, Goldberg MS, Valois MF, Nadon L. Postmenopausal breast cancer and occupational exposures. Occup Environ Med. 2010;67(4):263-269. doi:10.1136/oem.2009.049817

55. National Institute of Environmental Health Sciences, Interagency Breast Cancer & Environmental Research Coordinating Committee. Breast cancer and the environment: prioritizing prevention. Updated March 8, 2013. Accessed April 12, 2021. https://www.niehs.nih.gov/about/boards/ibcercc/index.cfm

56. Gail MH, Costantino JP, Pee D, et al. Projecting individualized absolute invasive breast cancer risk in African American women [published correction appears in J Natl Cancer Inst. 2008 Aug 6;100(15):1118] [published correction appears in J Natl Cancer Inst. 2008 Mar 5;100(5):373]. J Natl Cancer Inst. 2007;99(23):1782-1792. doi:10.1093/jnci/djm223

57. Corbie-Smith G, Thomas SB, Williams MV, Moody-Ayers S. Attitudes and beliefs of African Americans toward participation in medical research. J Gen Intern Med. 1999;14(9):537-546. doi:10.1046/j.1525-1497.1999.07048.x

58. Braunstein JB, Sherber NS, Schulman SP, Ding EL, Powe NR. Race, medical researcher distrust, perceived harm, and willingness to participate in cardiovascular prevention trials. Medicine (Baltimore). 2008;87(1):1-9. doi:10.1097/MD.0b013e3181625d78

References

1. US Department of Veterans Affairs. National Center for Veterans Analysis and Statistics. The past, present and future of women veterans. Published February 2017. Accessed April 28, 2021. https://www.va.gov/vetdata/docs/specialreports/women_veterans_2015_final.pdf.

2. Frayne SM, Carney DV, Bastian L, et al. The VA Women’s Health Practice-Based Research Network: amplifying women veterans’ voices in VA research. J Gen Intern Med. 2013;28 Suppl 2(Suppl 2):S504-S509. doi:10.1007/s11606-013-2476-3

3. US Department of Veterans Affairs, Veterans Health Administration, Women’s Health Evaluation Initiative, Women Veterans Health Strategic Health Care Group. Sourcebook: women veterans in the Veterans Health Administration. Volume 1: Sociodemographic characteristics and use of VHA care. Published December 2010. Accessed April 12, 2021. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=2455

4. Bean-Mayberry B, Yano EM, Bayliss N, Navratil J, Weisman CS, Scholle SH. Federally funded comprehensive women’s health centers: leading innovation in women’s healthcare delivery. J Womens Health (Larchmt). 2007;16(9):1281-1290. doi:10.1089/jwh.2006.0284

5. US Department of Veterans Affairs. National Center for Veterans Analysis and Statistics.VA utilization profile FY 2016. Published November 2017. Accessed April 12, 2021. https://www.va.gov/vetdata/docs/QuickFacts/VA_Utilization_Profile.PDF

6. Ekenga CC, Parks CG, Sandler DP. Chemical exposures in the workplace and breast cancer risk: a prospective cohort study. Int J Cancer. 2015;137(7):1765-1774. doi:10.1002/ijc.29545

7. Rennix CP, Quinn MM, Amoroso PJ, Eisen EA, Wegman DH. Risk of breast cancer among enlisted Army women occupationally exposed to volatile organic compounds. Am J Ind Med. 2005;48(3):157-167. doi:10.1002/ajim.20201

8. Ritz B. Cancer mortality among workers exposed to chemicals during uranium processing. J Occup Environ Med. 1999;41(7):556-566. doi:10.1097/00043764-199907000-00004

9. Zhu K, Devesa SS, Wu H, et al. Cancer incidence in the U.S. military population: comparison with rates from the SEER program. Cancer Epidemiol Biomarkers Prev. 2009;18(6):1740-1745. doi:10.1158/1055-9965.EPI-09-0041

10. Freedman AN, Yu B, Gail MH, et al. Benefit/risk assessment for breast cancer chemoprevention with raloxifene or tamoxifen for women age 50 years or older [published correction appears in J Clin Oncol. 2013 Nov 10;31(32):4167]. J Clin Oncol. 2011;29(17):2327-2333. doi:10.1200/JCO.2010.33.0258

11. Greene, H. Cancer prevention, screening and early detection. In: Gobel BH, Triest-Robertson S, Vogel WH, eds. Advanced Oncology Nursing Certification Review and Resource Manual. 3rd ed. Oncology Nursing Society; 2016:1-34. https://www.ons.org/sites/default/files/publication_pdfs/2%20ADVPrac%20chapter%201.pdf

12. National Comprehensive Cancer Network. NCCN Breast Cancer Risk Reduction. Version 1.2021 NCCN Clinical Practice Guidelines in Oncology. Updated March 24, 2021 Accessed April 12, 2021. https://www.nccn.org/professionals/physician_gls/pdf/breast_risk.pdf

13. US Preventive Services Task Force. Breast cancer: Medications use to reduce risk. Updated September 3, 2019. Accessed April 12, 2021. https://www.uspreventiveservicestaskforce.org/uspstf/recommendation/breast-cancer-medications-for-risk-reduction

14. Moyer VA; U.S. Preventive Services Task Force. Medications to decrease the risk for breast cancer in women: recommendations from the U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2013;159(10):698-708. doi:10.7326/0003-4819-159-10-201311190-00717

15. Boucher JE. Chemoprevention: an overview of pharmacologic agents and nursing considerations. Clin J Oncol Nurs. 2018;22(3):350-353. doi:10.1188/18.CJON.350-353

16. Nichols HB, Stürmer T, Lee VS, et al. Breast cancer chemoprevention in an integrated health care setting. JCO Clin Cancer Inform. 2017;1:1-12. doi:10.1200/CCI.16.00059

17. Bevers TB, Helvie M, Bonaccio E, et al. Breast cancer screening and diagnosis, Version 3.2018, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2018;16(11):1362-1389. doi:10.6004/jnccn.2018.0083

18. Visvanathan K, Hurley P, Bantug E, et al. Use of pharmacologic interventions for breast cancer risk reduction: American Society of Clinical Oncology clinical practice guideline [published correction appears in J Clin Oncol. 2013 Dec 1;31(34):4383]. J Clin Oncol. 2013;31(23):2942-2962. doi:10.1200/JCO.2013.49.3122

19. Sealy-Jefferson S, Roseland ME, Cote ML, et al. rural-urban residence and stage at breast cancer diagnosis among postmenopausal women: The Women’s Health Initiative. J Womens Health (Larchmt). 2019;28(2):276-283. doi:10.1089/jwh.2017.6884

20. Holder KA. Veterans in rural America: 2011-2015. Published January 25, 2017. Accessed April 12, 2021. https://www.census.gov/library/publications/2017/acs/acs-36.html

21. Owens WL, Gallagher TJ, Kincheloe MJ, Ruetten VL. Implementation in a large health system of a program to identify women at high risk for breast cancer. J Oncol Pract. 2011;7(2):85-88. doi:10.1200/JOP.2010.000107

2. Pivot X, Viguier J, Touboul C, et al. Breast cancer screening controversy: too much or not enough?. Eur J Cancer Prev. 2015;24 Suppl:S73-S76. doi:10.1097/CEJ.0000000000000145

23. Bidassie B, Kovach A, Vallette MA, et al. Breast Cancer risk assessment and chemoprevention use among veterans affairs primary care providers: a national online survey. Mil Med. 2020;185(3-4):512-518. doi:10.1093/milmed/usz291

24. Brewster AM, Davidson NE, McCaskill-Stevens W. Chemoprevention for breast cancer: overcoming barriers to treatment. Am Soc Clin Oncol Educ Book. 2012;85-90. doi:10.14694/EdBook_AM.2012.32.152

25. Meyskens FL Jr, Curt GA, Brenner DE, et al. Regulatory approval of cancer risk-reducing (chemopreventive) drugs: moving what we have learned into the clinic. Cancer Prev Res (Phila). 2011;4(3):311-323. doi:10.1158/1940-6207.CAPR-09-0014

26. Tice JA, Kerlikowske K. Screening and prevention of breast cancer in primary care. Prim Care. 2009;36(3):533-558. doi:10.1016/j.pop.2009.04.003

27. Vogel VG. Selective estrogen receptor modulators and aromatase inhibitors for breast cancer chemoprevention. Curr Drug Targets. 2011;12(13):1874-1887. doi:10.2174/138945011798184164

28. Vogel VG, Costantino JP, Wickerham DL, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial [published correction appears in JAMA. 2006 Dec 27;296(24):2926] [published correction appears in JAMA. 2007 Sep 5;298(9):973]. JAMA. 2006;295(23):2727-2741. doi:10.1001/jama.295.23.joc60074

29. Pruthi S, Heisey RE, Bevers TB. Chemoprevention for breast cancer. Ann Surg Oncol. 2015;22(10):3230-3235. doi:10.1245/s10434-015-4715-9

30. Cuzick J, Sestak I, Forbes JF, et al. Anastrozole for prevention of breast cancer in high-risk postmenopausal women (IBIS-II): an international, double-blind, randomised placebo-controlled trial [published correction appears in Lancet. 2014 Mar 22;383(9922):1040] [published correction appears in Lancet. 2017 Mar 11;389(10073):1010]. Lancet. 2014;383(9922):1041-1048. doi:10.1016/S0140-6736(13)62292-8

31. Bozovic-Spasojevic I, Azambuja E, McCaskill-Stevens W, Dinh P, Cardoso F. Chemoprevention for breast cancer. Cancer Treat Rev. 2012;38(5):329-339. doi:10.1016/j.ctrv.2011.07.005

32. Gabriel EM, Jatoi I. Breast cancer chemoprevention. Expert Rev Anticancer Ther. 2012;12(2):223-228. doi:10.1586/era.11.206

<--pagebreak-->

33. Crew KD, Albain KS, Hershman DL, Unger JM, Lo SS. How do we increase uptake of tamoxifen and other anti-estrogens for breast cancer prevention?. NPJ Breast Cancer. 2017;3:20. Published 2017 May 19. doi:10.1038/s41523-017-0021-y

34. Ropka ME, Keim J, Philbrick JT. Patient decisions about breast cancer chemoprevention: a systematic review and meta-analysis. J Clin Oncol. 2010;28(18):3090-3095. doi:10.1200/JCO.2009.27.8077

35. Smith SG, Sestak I, Forster A, et al. Factors affecting uptake and adherence to breast cancer chemoprevention: a systematic review and meta-analysis. Ann Oncol. 2016;27(4):575-590. doi:10.1093/annonc/mdv590

36. Grann VR, Patel PR, Jacobson JS, et al. Comparative effectiveness of screening and prevention strategies among BRCA1/2-affected mutation carriers. Breast Cancer Res Treat. 2011 Feb;125(3):837-847. doi:10.1007/s10549-010-1043-4

37. Goss PE, Ingle JN, Alés-Martínez JE, et al. Exemestane for breast-cancer prevention in postmenopausal women [published correction appears in N Engl J Med. 2011 Oct 6;365(14):1361]. N Engl J Med. 2011;364(25):2381-2391. doi:10.1056/NEJMoa1103507

38. Kmietowicz Z. Five in six women reject drugs that could reduce their risk of breast cancer. BMJ. 2015;351:h6650. Published 2015 Dec 8. doi:10.1136/bmj.h6650

39. Nelson HD, Fu R, Griffin JC, Nygren P, Smith ME, Humphrey L. Systematic review: comparative effectiveness of medications to reduce risk for primary breast cancer. Ann Intern Med. 2009;151(10):703-235. doi:10.7326/0003-4819-151-10-200911170-00147

40. Dahabreh IJ, Wieland LS, Adam GP, Halladay C, Lau J, Trikalinos TA. Core needle and open surgery biopsy for diagnosis of breast lesions: an update to the 2009 report. Published September 2014. Accessed April 12, 2021. https://www.ncbi.nlm.nih.gov/books/NBK246878

41. National Cancer Institute. Genetics of breast and ovarian cancer (PDQ)—health profession version. Updated February 12, 2021. Accessed April 12, 2021. http://www.cancer.gov/cancertopics/pdq/genetics/breast-and-ovarian/HealthProfessional

42. US Department of Health and Human Services. National Institutes of Health, National Institute of Environmental Health Sciences The sister study. Accessed April 12, 2021. https://sisterstudy.niehs.nih.gov/english/NIEHS.htm

43. Tutt A, Ashworth A. Can genetic testing guide treatment in breast cancer?. Eur J Cancer. 2008;44(18):2774-2780. doi:10.1016/j.ejca.2008.10.009

44. Katz SJ, Ward KC, Hamilton AS, et al. Gaps in receipt of clinically indicated genetic counseling after diagnosis of breast cancer. J Clin Oncol. 2018;36(12):1218-1224. doi:10.1200/JCO.2017.76.2369

45. US Department of Veterans Affairs. PTSD: National Center for PTSD. How common is PTSD in adults? Updated October 17, 2019. Accessed April 12, 2021. https://www.ptsd.va.gov/understand/common/common_adults.asp

46. US Department of Veterans Affairs. PTSD: National Center for PTSD. How common is PTSD in women? Updated October 16, 2019. Accessed April 12, 2021. https://www.ptsd.va.gov/understand/common/common_women.asp

47. US Department of Veterans Affairs. PTSD: National Center for PTSD. How common is PTSD in veterans? Updated September 24, 2018. Accessed April 12, 2021. https://www.ptsd.va.gov/understand/common/common_veterans.asp

48. Lindberg NM, Wellisch D. Anxiety and compliance among women at high risk for breast cancer. Ann Behav Med. 2001;23(4):298-303. doi:10.1207/S15324796ABM2304_9

49. DiMatteo MR, Lepper HS, Croghan TW. Depression is a risk factor for noncompliance with medical treatment: meta-analysis of the effects of anxiety and depression on patient adherence. Arch Intern Med. 2000;160(14):2101-2107. doi:10.1001/archinte.160.14.2101

50. Centers for Disease Control and Prevention. MMWR appendix: breast cancer rates among black women and white women. Updated October 13, 2016. Accessed April 12, 2021. https://www.cdc.gov/cancer/breast/statistics/trends_invasive.htm

51. Richardson LC, Henley SJ, Miller JW, Massetti G, Thomas CC. Patterns and trends in age-specific black-white differences in breast cancer incidence and mortality - United States, 1999-2014. MMWR Morb Mortal Wkly Rep. 2016;65(40):1093-1098. Published 2016 Oct 14. doi:10.15585/mmwr.mm6540a1

52. Brody JG, Moysich KB, Humblet O, Attfield KR, Beehler GP, Rudel RA. Environmental pollutants and breast cancer: epidemiologic studies. Cancer. 2007;109(12 Suppl):2667-2711. doi:10.1002/cncr.22655

53. Brophy JT, Keith MM, Watterson A, et al. Breast cancer risk in relation to occupations with exposure to carcinogens and endocrine disruptors: a Canadian case-control study. Environ Health. 2012;11:87. Published 2012 Nov 19. doi:10.1186/1476-069X-11-87

54. Labrèche F, Goldberg MS, Valois MF, Nadon L. Postmenopausal breast cancer and occupational exposures. Occup Environ Med. 2010;67(4):263-269. doi:10.1136/oem.2009.049817

55. National Institute of Environmental Health Sciences, Interagency Breast Cancer & Environmental Research Coordinating Committee. Breast cancer and the environment: prioritizing prevention. Updated March 8, 2013. Accessed April 12, 2021. https://www.niehs.nih.gov/about/boards/ibcercc/index.cfm

56. Gail MH, Costantino JP, Pee D, et al. Projecting individualized absolute invasive breast cancer risk in African American women [published correction appears in J Natl Cancer Inst. 2008 Aug 6;100(15):1118] [published correction appears in J Natl Cancer Inst. 2008 Mar 5;100(5):373]. J Natl Cancer Inst. 2007;99(23):1782-1792. doi:10.1093/jnci/djm223

57. Corbie-Smith G, Thomas SB, Williams MV, Moody-Ayers S. Attitudes and beliefs of African Americans toward participation in medical research. J Gen Intern Med. 1999;14(9):537-546. doi:10.1046/j.1525-1497.1999.07048.x

58. Braunstein JB, Sherber NS, Schulman SP, Ding EL, Powe NR. Race, medical researcher distrust, perceived harm, and willingness to participate in cardiovascular prevention trials. Medicine (Baltimore). 2008;87(1):1-9. doi:10.1097/MD.0b013e3181625d78

Issue
Federal Practitioner - 38(2)s
Issue
Federal Practitioner - 38(2)s
Page Number
S35-S41
Page Number
S35-S41
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
Article PDF Media

Small increase seen in new COVID-19 cases among children

Article Type
Changed
Thu, 08/26/2021 - 15:47

 

After 2 consecutive weeks of declines, the number of new COVID-19 cases in children rose slightly, according to a report from the American Academy of Pediatrics and the Children’s Hospital Association.

The increase was less than 0.6% – 72,067 cases were reported during the week of April 30 to May 6, compared with 71,649 a week earlier – but when combined with a drop in new cases overall it brought children’s share of all COVID-19 cases for the week to 24.0%, higher than at any other time during the pandemic, the AAP and CHA data show.

It is worth noting, however, that Rhode Island experienced a 30% increase in the last week, adding about 4,900 cases because of data revision and a lag in reporting, the AAP and CHA said in their weekly COVID-19 report.

All the new cases bring the total national count to just over 3.54 million in children, which represents 14.0% of all cases in 49 states (excluding New York), the District of Columbia, New York City, Puerto Rico, and Guam. The cumulative case rate as of May 6 was 5,122 per 100,000 children, the two organizations said.



All the new cases that were added to Rhode Island’s total give it the highest cumulative rate in the country: 9,614 cases per 100,000 children. North Dakota is right behind with 9,526 per 100,000, followed by Tennessee (8,898), Connecticut (8,281), and South Carolina (8,274). Vermont has the highest proportion of cases in children at 22.4%, with Alaska next at 20.3% and South Carolina third at 18.7%, according to the AAP and CHA.

Hawaii just reported its first COVID-19–related death in a child, which drops the number of states with zero deaths in children from 10 to 9. Two other new deaths in children from April 30 to May 6 bring the total number to 306 in the 43 states, along with New York City, Puerto Rico, and Guam, that are reporting the age distribution of deaths.

In a separate statement, AAP president Lee Savio Beers acknowledged the Food and Drug Administration’s authorization of the Pfizer-BioNTech vaccine for children aged 12-15 years as “a critically important step in bringing lifesaving vaccines to children and adolescents. ... We look forward to the discussion by the Advisory Committee on Immunization Practices of the CDC, which will make recommendations about the use of this vaccine in adolescents.”

Publications
Topics
Sections

 

After 2 consecutive weeks of declines, the number of new COVID-19 cases in children rose slightly, according to a report from the American Academy of Pediatrics and the Children’s Hospital Association.

The increase was less than 0.6% – 72,067 cases were reported during the week of April 30 to May 6, compared with 71,649 a week earlier – but when combined with a drop in new cases overall it brought children’s share of all COVID-19 cases for the week to 24.0%, higher than at any other time during the pandemic, the AAP and CHA data show.

It is worth noting, however, that Rhode Island experienced a 30% increase in the last week, adding about 4,900 cases because of data revision and a lag in reporting, the AAP and CHA said in their weekly COVID-19 report.

All the new cases bring the total national count to just over 3.54 million in children, which represents 14.0% of all cases in 49 states (excluding New York), the District of Columbia, New York City, Puerto Rico, and Guam. The cumulative case rate as of May 6 was 5,122 per 100,000 children, the two organizations said.



All the new cases that were added to Rhode Island’s total give it the highest cumulative rate in the country: 9,614 cases per 100,000 children. North Dakota is right behind with 9,526 per 100,000, followed by Tennessee (8,898), Connecticut (8,281), and South Carolina (8,274). Vermont has the highest proportion of cases in children at 22.4%, with Alaska next at 20.3% and South Carolina third at 18.7%, according to the AAP and CHA.

Hawaii just reported its first COVID-19–related death in a child, which drops the number of states with zero deaths in children from 10 to 9. Two other new deaths in children from April 30 to May 6 bring the total number to 306 in the 43 states, along with New York City, Puerto Rico, and Guam, that are reporting the age distribution of deaths.

In a separate statement, AAP president Lee Savio Beers acknowledged the Food and Drug Administration’s authorization of the Pfizer-BioNTech vaccine for children aged 12-15 years as “a critically important step in bringing lifesaving vaccines to children and adolescents. ... We look forward to the discussion by the Advisory Committee on Immunization Practices of the CDC, which will make recommendations about the use of this vaccine in adolescents.”

 

After 2 consecutive weeks of declines, the number of new COVID-19 cases in children rose slightly, according to a report from the American Academy of Pediatrics and the Children’s Hospital Association.

The increase was less than 0.6% – 72,067 cases were reported during the week of April 30 to May 6, compared with 71,649 a week earlier – but when combined with a drop in new cases overall it brought children’s share of all COVID-19 cases for the week to 24.0%, higher than at any other time during the pandemic, the AAP and CHA data show.

It is worth noting, however, that Rhode Island experienced a 30% increase in the last week, adding about 4,900 cases because of data revision and a lag in reporting, the AAP and CHA said in their weekly COVID-19 report.

All the new cases bring the total national count to just over 3.54 million in children, which represents 14.0% of all cases in 49 states (excluding New York), the District of Columbia, New York City, Puerto Rico, and Guam. The cumulative case rate as of May 6 was 5,122 per 100,000 children, the two organizations said.



All the new cases that were added to Rhode Island’s total give it the highest cumulative rate in the country: 9,614 cases per 100,000 children. North Dakota is right behind with 9,526 per 100,000, followed by Tennessee (8,898), Connecticut (8,281), and South Carolina (8,274). Vermont has the highest proportion of cases in children at 22.4%, with Alaska next at 20.3% and South Carolina third at 18.7%, according to the AAP and CHA.

Hawaii just reported its first COVID-19–related death in a child, which drops the number of states with zero deaths in children from 10 to 9. Two other new deaths in children from April 30 to May 6 bring the total number to 306 in the 43 states, along with New York City, Puerto Rico, and Guam, that are reporting the age distribution of deaths.

In a separate statement, AAP president Lee Savio Beers acknowledged the Food and Drug Administration’s authorization of the Pfizer-BioNTech vaccine for children aged 12-15 years as “a critically important step in bringing lifesaving vaccines to children and adolescents. ... We look forward to the discussion by the Advisory Committee on Immunization Practices of the CDC, which will make recommendations about the use of this vaccine in adolescents.”

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

Making the Rounds to Diagnosis

Article Type
Changed
Mon, 05/17/2021 - 10:28
Display Headline
Making the Rounds to Diagnosis

ANSWER

The correct answer is granuloma annulare (GA; choice “d”).

DISCUSSION

The biopsy showed rings of dermal epithelioid histiocytes surrounding a core of central mucin, configured in rows (the latter conferring the diagnostic term “palisaded” granulomas). Both the pathology results and the morphology of the lesions—color, shape, etc—were classic for GA. What was somewhat unusual about this case was the size and number of lesions, which are typically fewer and smaller with GA.

GA is seldom much of a problem and has no connection to serious disease. But it can, as this case illustrates, mimic some rather worrisome conditions. Had this been sarcoidosis (choice “a”), there would be no central “delling” (concavity), and the biopsy would have shown necrotic (caseating) nonpalisading granulomas.

Cutaneous T-cell lymphoma (CTCL; choice “b”) can manifest with plaques, but there wouldn’t be any delling and the shapes would not be so consistently round. CTCL will eventually give rise to palpable adenopathy.

Lupus profundus (choice “c”) also manifests with deep plaques, without delling. It is a truly rare variant of lupus, which would have been detected on biopsy.

Concern about these differential items is what drove the decision to perform full-thickness punch biopsies as opposed to taking a simple shave sample.

TREATMENT

The most common treatment for GA is intralesional steroid injection (eg, 10 mg/cc triamcinolone) of large lesions. Oral medications such as methotrexate or pentoxifylline have been used with some success. Often, the condition is self-limiting.

Author and Disclosure Information

Joe R. Monroe, MPAS, PA, practices at Dermatology Associates of Oklahoma in Tulsa. He is also the founder of the Society of Dermatology Physician Assistants.

Publications
Topics
Sections
Author and Disclosure Information

Joe R. Monroe, MPAS, PA, practices at Dermatology Associates of Oklahoma in Tulsa. He is also the founder of the Society of Dermatology Physician Assistants.

Author and Disclosure Information

Joe R. Monroe, MPAS, PA, practices at Dermatology Associates of Oklahoma in Tulsa. He is also the founder of the Society of Dermatology Physician Assistants.

ANSWER

The correct answer is granuloma annulare (GA; choice “d”).

DISCUSSION

The biopsy showed rings of dermal epithelioid histiocytes surrounding a core of central mucin, configured in rows (the latter conferring the diagnostic term “palisaded” granulomas). Both the pathology results and the morphology of the lesions—color, shape, etc—were classic for GA. What was somewhat unusual about this case was the size and number of lesions, which are typically fewer and smaller with GA.

GA is seldom much of a problem and has no connection to serious disease. But it can, as this case illustrates, mimic some rather worrisome conditions. Had this been sarcoidosis (choice “a”), there would be no central “delling” (concavity), and the biopsy would have shown necrotic (caseating) nonpalisading granulomas.

Cutaneous T-cell lymphoma (CTCL; choice “b”) can manifest with plaques, but there wouldn’t be any delling and the shapes would not be so consistently round. CTCL will eventually give rise to palpable adenopathy.

Lupus profundus (choice “c”) also manifests with deep plaques, without delling. It is a truly rare variant of lupus, which would have been detected on biopsy.

Concern about these differential items is what drove the decision to perform full-thickness punch biopsies as opposed to taking a simple shave sample.

TREATMENT

The most common treatment for GA is intralesional steroid injection (eg, 10 mg/cc triamcinolone) of large lesions. Oral medications such as methotrexate or pentoxifylline have been used with some success. Often, the condition is self-limiting.

ANSWER

The correct answer is granuloma annulare (GA; choice “d”).

DISCUSSION

The biopsy showed rings of dermal epithelioid histiocytes surrounding a core of central mucin, configured in rows (the latter conferring the diagnostic term “palisaded” granulomas). Both the pathology results and the morphology of the lesions—color, shape, etc—were classic for GA. What was somewhat unusual about this case was the size and number of lesions, which are typically fewer and smaller with GA.

GA is seldom much of a problem and has no connection to serious disease. But it can, as this case illustrates, mimic some rather worrisome conditions. Had this been sarcoidosis (choice “a”), there would be no central “delling” (concavity), and the biopsy would have shown necrotic (caseating) nonpalisading granulomas.

Cutaneous T-cell lymphoma (CTCL; choice “b”) can manifest with plaques, but there wouldn’t be any delling and the shapes would not be so consistently round. CTCL will eventually give rise to palpable adenopathy.

Lupus profundus (choice “c”) also manifests with deep plaques, without delling. It is a truly rare variant of lupus, which would have been detected on biopsy.

Concern about these differential items is what drove the decision to perform full-thickness punch biopsies as opposed to taking a simple shave sample.

TREATMENT

The most common treatment for GA is intralesional steroid injection (eg, 10 mg/cc triamcinolone) of large lesions. Oral medications such as methotrexate or pentoxifylline have been used with some success. Often, the condition is self-limiting.

Publications
Publications
Topics
Article Type
Display Headline
Making the Rounds to Diagnosis
Display Headline
Making the Rounds to Diagnosis
Sections
Questionnaire Body

Lesion

A 60-year-old woman is feeling fine—no fever, arthralgia, or malaise—but the sudden appearance of lesions on her extremities unsettles her to the point that she can think of little else. Visits to her primary care provider, the emergency department, several urgent care clinics, and a naturopath yield a consistent diagnosis: ringworm. Yet none of the topical or oral antifungal medications she is prescribed make the slightest difference. Thus, she finally agrees to consult a dermatologist.

The patient reports decent health, although she was a heavy smoker for years before quitting 2 years ago. She recently tested negative for diabetes.

History taking reveals no sources from which she could have acquired a fungal infection. No one else at home is similarly affected.

About 10 lesions, mostly located on the patient’s extremities, are examined. All are very similar in appearance: round, brownish-red intradermal plaques with no epidermal component (eg, scale, broken skin). The lesions vary from 6 mm to 4 cm in diameter. The centers of most lesions are slightly concave, with well-defined raised margins. On palpation, there is neither increased warmth nor tenderness. No nodes can be palpated in the groin, axillae, or epitrochlear locations.

A deep punch biopsy is performed on a thigh lesion, with the specimen submitted for pathologic examination.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 05/11/2021 - 14:00
Un-Gate On Date
Tue, 05/11/2021 - 14:00
Use ProPublica
CFC Schedule Remove Status
Tue, 05/11/2021 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Dr. Fauci: Feds may ease indoor mask mandates soon

Article Type
Changed
Thu, 08/26/2021 - 15:47

 

Federal guidance on indoor mask use may change soon, Anthony S. Fauci, MD, director of the National Institute of Allergy and Infectious Diseases, said on May 9.

He was asked whether it’s time to start relaxing indoor mask requirements.

“I think so, and I think you’re going to probably be seeing that as we go along and as more people get vaccinated,” Dr. Fauci said on ABC News’s This Week.Nearly 150 million adults in the United States – or about 58% of the adult population – have received at least one COVID-19 vaccine dose, according to the latest CDC tally. About 113 million adults, or 44%, are considered fully vaccinated.

“The CDC will be, you know, almost in real time … updating their recommendations and their guidelines,” Dr. Fauci said.

In April, the CDC relaxed its guidance for those who have been vaccinated against COVID-19. Those who have gotten a shot don’t need to wear a mask outdoors or in small indoor gatherings with other vaccinated people, but both vaccinated and unvaccinated people are still advised to wear masks in indoor public spaces.

“We do need to start being more liberal as we get more people vaccinated,” Dr. Fauci said. “As you get more people vaccinated, the number of cases per day will absolutely go down.”

The United States is averaging about 43,000 cases per day, he said, adding that the cases need to be “much, much lower.” When the case numbers drop and vaccination numbers increase, the risk of infection will fall dramatically indoors and outdoors, he said.

Even after the pandemic, though, wearing masks could become a seasonal habit, Dr. Fauci said May 9 on NBC News’s Meet the Press.“I think people have gotten used to the fact that wearing masks, clearly if you look at the data, it diminishes respiratory diseases. We’ve had practically a nonexistent flu season this year,” he said.

“So it is conceivable that as we go on, a year or 2 or more from now, that during certain seasonal periods when you have respiratory-borne viruses like the flu, people might actually elect to wear masks to diminish the likelihood that you’ll spread these respiratory-borne diseases,” he said.

Dr. Fauci was asked about indoor mask guidelines on May 9 after former FDA Commissioner Scott Gottlieb, MD, said face mask requirements should be relaxed.

“Certainly outdoors, we shouldn’t be putting limits on gatherings anymore,” Dr. Gottlieb said on CBS News’s Face the Nation.“The states where prevalence is low, vaccination rates are high, we have good testing in place, and we’re identifying infections, I think we could start lifting these restrictions indoors as well, on a broad basis,” he said.

Lifting pandemic-related restrictions in areas where they’re no longer necessary could also encourage people to implement them again if cases increase during future surges, such as this fall or winter, Dr. Gottlieb said.

At the same time, Americans should continue to follow CDC guidance and wait for new guidelines before changing their indoor mask use, Jeffrey Zients, the White House COVID-19 response coordinator, said on CNN’s State of the Union on May 9.

“We all want to get back to a normal lifestyle,” he said. “I think we’re on the path to do that, but stay disciplined, and let’s take advantage of the new privilege of being vaccinated and not wearing masks outdoors, for example, unless you’re in a crowded place.”

Mr. Zients pointed to President Joe Biden’s goal for 70% of adults to receive at least one vaccine dose by July 4.

“As we all move toward that 70% goal, there will be more and more advantages to being vaccinated,” he said. “And if you’re not vaccinated, you’re not protected.”

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

Publications
Topics
Sections

 

Federal guidance on indoor mask use may change soon, Anthony S. Fauci, MD, director of the National Institute of Allergy and Infectious Diseases, said on May 9.

He was asked whether it’s time to start relaxing indoor mask requirements.

“I think so, and I think you’re going to probably be seeing that as we go along and as more people get vaccinated,” Dr. Fauci said on ABC News’s This Week.Nearly 150 million adults in the United States – or about 58% of the adult population – have received at least one COVID-19 vaccine dose, according to the latest CDC tally. About 113 million adults, or 44%, are considered fully vaccinated.

“The CDC will be, you know, almost in real time … updating their recommendations and their guidelines,” Dr. Fauci said.

In April, the CDC relaxed its guidance for those who have been vaccinated against COVID-19. Those who have gotten a shot don’t need to wear a mask outdoors or in small indoor gatherings with other vaccinated people, but both vaccinated and unvaccinated people are still advised to wear masks in indoor public spaces.

“We do need to start being more liberal as we get more people vaccinated,” Dr. Fauci said. “As you get more people vaccinated, the number of cases per day will absolutely go down.”

The United States is averaging about 43,000 cases per day, he said, adding that the cases need to be “much, much lower.” When the case numbers drop and vaccination numbers increase, the risk of infection will fall dramatically indoors and outdoors, he said.

Even after the pandemic, though, wearing masks could become a seasonal habit, Dr. Fauci said May 9 on NBC News’s Meet the Press.“I think people have gotten used to the fact that wearing masks, clearly if you look at the data, it diminishes respiratory diseases. We’ve had practically a nonexistent flu season this year,” he said.

“So it is conceivable that as we go on, a year or 2 or more from now, that during certain seasonal periods when you have respiratory-borne viruses like the flu, people might actually elect to wear masks to diminish the likelihood that you’ll spread these respiratory-borne diseases,” he said.

Dr. Fauci was asked about indoor mask guidelines on May 9 after former FDA Commissioner Scott Gottlieb, MD, said face mask requirements should be relaxed.

“Certainly outdoors, we shouldn’t be putting limits on gatherings anymore,” Dr. Gottlieb said on CBS News’s Face the Nation.“The states where prevalence is low, vaccination rates are high, we have good testing in place, and we’re identifying infections, I think we could start lifting these restrictions indoors as well, on a broad basis,” he said.

Lifting pandemic-related restrictions in areas where they’re no longer necessary could also encourage people to implement them again if cases increase during future surges, such as this fall or winter, Dr. Gottlieb said.

At the same time, Americans should continue to follow CDC guidance and wait for new guidelines before changing their indoor mask use, Jeffrey Zients, the White House COVID-19 response coordinator, said on CNN’s State of the Union on May 9.

“We all want to get back to a normal lifestyle,” he said. “I think we’re on the path to do that, but stay disciplined, and let’s take advantage of the new privilege of being vaccinated and not wearing masks outdoors, for example, unless you’re in a crowded place.”

Mr. Zients pointed to President Joe Biden’s goal for 70% of adults to receive at least one vaccine dose by July 4.

“As we all move toward that 70% goal, there will be more and more advantages to being vaccinated,” he said. “And if you’re not vaccinated, you’re not protected.”

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

 

Federal guidance on indoor mask use may change soon, Anthony S. Fauci, MD, director of the National Institute of Allergy and Infectious Diseases, said on May 9.

He was asked whether it’s time to start relaxing indoor mask requirements.

“I think so, and I think you’re going to probably be seeing that as we go along and as more people get vaccinated,” Dr. Fauci said on ABC News’s This Week.Nearly 150 million adults in the United States – or about 58% of the adult population – have received at least one COVID-19 vaccine dose, according to the latest CDC tally. About 113 million adults, or 44%, are considered fully vaccinated.

“The CDC will be, you know, almost in real time … updating their recommendations and their guidelines,” Dr. Fauci said.

In April, the CDC relaxed its guidance for those who have been vaccinated against COVID-19. Those who have gotten a shot don’t need to wear a mask outdoors or in small indoor gatherings with other vaccinated people, but both vaccinated and unvaccinated people are still advised to wear masks in indoor public spaces.

“We do need to start being more liberal as we get more people vaccinated,” Dr. Fauci said. “As you get more people vaccinated, the number of cases per day will absolutely go down.”

The United States is averaging about 43,000 cases per day, he said, adding that the cases need to be “much, much lower.” When the case numbers drop and vaccination numbers increase, the risk of infection will fall dramatically indoors and outdoors, he said.

Even after the pandemic, though, wearing masks could become a seasonal habit, Dr. Fauci said May 9 on NBC News’s Meet the Press.“I think people have gotten used to the fact that wearing masks, clearly if you look at the data, it diminishes respiratory diseases. We’ve had practically a nonexistent flu season this year,” he said.

“So it is conceivable that as we go on, a year or 2 or more from now, that during certain seasonal periods when you have respiratory-borne viruses like the flu, people might actually elect to wear masks to diminish the likelihood that you’ll spread these respiratory-borne diseases,” he said.

Dr. Fauci was asked about indoor mask guidelines on May 9 after former FDA Commissioner Scott Gottlieb, MD, said face mask requirements should be relaxed.

“Certainly outdoors, we shouldn’t be putting limits on gatherings anymore,” Dr. Gottlieb said on CBS News’s Face the Nation.“The states where prevalence is low, vaccination rates are high, we have good testing in place, and we’re identifying infections, I think we could start lifting these restrictions indoors as well, on a broad basis,” he said.

Lifting pandemic-related restrictions in areas where they’re no longer necessary could also encourage people to implement them again if cases increase during future surges, such as this fall or winter, Dr. Gottlieb said.

At the same time, Americans should continue to follow CDC guidance and wait for new guidelines before changing their indoor mask use, Jeffrey Zients, the White House COVID-19 response coordinator, said on CNN’s State of the Union on May 9.

“We all want to get back to a normal lifestyle,” he said. “I think we’re on the path to do that, but stay disciplined, and let’s take advantage of the new privilege of being vaccinated and not wearing masks outdoors, for example, unless you’re in a crowded place.”

Mr. Zients pointed to President Joe Biden’s goal for 70% of adults to receive at least one vaccine dose by July 4.

“As we all move toward that 70% goal, there will be more and more advantages to being vaccinated,” he said. “And if you’re not vaccinated, you’re not protected.”

A version of this article first appeared on WebMD.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

Suicide risk prediction tools fail people of color

Article Type
Changed
Tue, 05/11/2021 - 12:54

Current models used to predict suicide risk fall short for racialized populations including Black, Indigenous, and people of color (BIPOC), new research shows.

Investigators developed two suicide prediction models to examine whether these types of tools are accurate in their predictive abilities, or whether they are flawed.

They found both prediction models failed to identify high-risk BIPOC individuals. In the first model, nearly half of outpatient visits followed by suicide were identified in White patients versus only 7% of visits followed by suicide in BIPOC patients. The second model had a sensitivity of 41% for White patients, but just 3% for Black patients and 7% for American Indian/Alaskan Native patients.

Dr. Yates Coley


“You don’t know whether a prediction model will be useful or harmful until it’s evaluated. The take-home message of our study is this: You have to look,” lead author Yates Coley, PhD, assistant investigator, Kaiser Permanente Washington Health Research Institute, Seattle, said in an interview.

The study was published online April 28, 2021, in JAMA Psychiatry.

Racial inequities

Suicide risk prediction models have been “developed and validated in several settings” and are now in regular use at the Veterans Health Administration, HealthPartners, and Kaiser Permanente, the authors wrote.

But the performance of suicide risk prediction models, while accurate in the overall population, “remains unexamined” in particular subpopulations, they noted.

“Health records data reflect existing racial and ethnic inequities in health care access, quality, and outcomes; and prediction models using health records data may perpetuate these disparities by presuming that past healthcare patterns accurately reflect actual needs,” Dr. Coley said.

Dr. Coley and associates “wanted to make sure that any suicide prediction model we implemented in clinical care reduced health disparities rather than exacerbated them.”

To investigate, researchers examined all outpatient mental health visits to seven large integrated health care systems by patients 13 years and older (n = 13,980,570 visits by 1,422,534 patients; 64% female, mean age, 42 years). The study spanned from Jan. 1, 2009, to Sept. 30, 2017, with follow-up through Dec. 31, 2017.

In particular, researchers looked at suicides that took place within 90 days following the outpatient visit.

Researchers used two prediction models: logistic regression with LASSO (Least Absolute Shrinkage and Selection Operator) variable selection and random forest technique, a “tree-based method that explores interactions between predictors (including those with race and ethnicity) in estimating probability of an outcome.”

The models considered prespecified interactions between predictors, including prior diagnoses, suicide attempts, and PHQ-9 [Patient Health Questionnaire–9] responses, and race and ethnicity data.

Researchers evaluated performance of the prediction models in the overall validation set and within subgroups defined by race/ethnicity.

The area under the curve measured model discrimination, and sensitivity was estimated for global and race/ethnicity-specific thresholds.
 

‘Unacceptable’ scenario

Within the total population, there were 768 deaths by suicide within 90 days of 3,143 visits. Suicide rates were highest for visits by patients with no recorded race/ethnicity, followed by visits by Asian, White, American Indian/Alaskan Native, Hispanic, and Black patients.

Both models showed “high” AUC sensitivity for White, Hispanic, and Asian patients but “poor” AUC sensitivity for BIPOC and patients without recorded race/ethnicity, the authors reported.

“Implementation of prediction models has to be considered in the broader context of unmet health care needs,” said Dr. Coley.

“In our specific example of suicide prediction, BIPOC populations already face substantial barriers in accessing quality mental health care and, as a result, have poorer outcomes, and using either of the suicide prediction models examined in our study will provide less benefit to already-underserved populations and widen existing care gaps,” a scenario Dr. Coley said is “unacceptable.”

We must insist that new technologies and methods be used to reduce racial and ethnic inequities in care, not exacerbate them,” she added.
 

 

 

Biased algorithms

Commenting on the study, Jonathan Singer, PhD, LCSW, associate professor at Loyola University, Chicago, described it as an “important contribution because it points to a systemic problem and also to the fact that the algorithms we create are biased, created by humans, and humans are biased.”

Although the study focused on the health care system, Dr. Singer believes the findings have implications for individual clinicians.

“If clinicians may be biased against identifying suicide risk in Black and Native American patients, they may attribute suicidal risk to something else. For example, we know that in Black Americans, expressions of intense emotions are oftentimes interpreted as aggression or being threatening, as opposed to indicators of sadness or fear,” noted Dr. Singer, who is also president of the American Academy of Suicidology and was not involved with the study,

“Clinicians who misinterpret these intense emotions are less likely to identify a Black client or patient who is suicidal,” Dr. Singer said.

The research was supported by the Mental Health Research Network from the National Institute of Mental Health. Dr. Coley has reported receiving support through a grant from the Agency for Healthcare Research and Quality. Dr. Singer reported no relevant financial relationships.

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

Publications
Topics
Sections

Current models used to predict suicide risk fall short for racialized populations including Black, Indigenous, and people of color (BIPOC), new research shows.

Investigators developed two suicide prediction models to examine whether these types of tools are accurate in their predictive abilities, or whether they are flawed.

They found both prediction models failed to identify high-risk BIPOC individuals. In the first model, nearly half of outpatient visits followed by suicide were identified in White patients versus only 7% of visits followed by suicide in BIPOC patients. The second model had a sensitivity of 41% for White patients, but just 3% for Black patients and 7% for American Indian/Alaskan Native patients.

Dr. Yates Coley


“You don’t know whether a prediction model will be useful or harmful until it’s evaluated. The take-home message of our study is this: You have to look,” lead author Yates Coley, PhD, assistant investigator, Kaiser Permanente Washington Health Research Institute, Seattle, said in an interview.

The study was published online April 28, 2021, in JAMA Psychiatry.

Racial inequities

Suicide risk prediction models have been “developed and validated in several settings” and are now in regular use at the Veterans Health Administration, HealthPartners, and Kaiser Permanente, the authors wrote.

But the performance of suicide risk prediction models, while accurate in the overall population, “remains unexamined” in particular subpopulations, they noted.

“Health records data reflect existing racial and ethnic inequities in health care access, quality, and outcomes; and prediction models using health records data may perpetuate these disparities by presuming that past healthcare patterns accurately reflect actual needs,” Dr. Coley said.

Dr. Coley and associates “wanted to make sure that any suicide prediction model we implemented in clinical care reduced health disparities rather than exacerbated them.”

To investigate, researchers examined all outpatient mental health visits to seven large integrated health care systems by patients 13 years and older (n = 13,980,570 visits by 1,422,534 patients; 64% female, mean age, 42 years). The study spanned from Jan. 1, 2009, to Sept. 30, 2017, with follow-up through Dec. 31, 2017.

In particular, researchers looked at suicides that took place within 90 days following the outpatient visit.

Researchers used two prediction models: logistic regression with LASSO (Least Absolute Shrinkage and Selection Operator) variable selection and random forest technique, a “tree-based method that explores interactions between predictors (including those with race and ethnicity) in estimating probability of an outcome.”

The models considered prespecified interactions between predictors, including prior diagnoses, suicide attempts, and PHQ-9 [Patient Health Questionnaire–9] responses, and race and ethnicity data.

Researchers evaluated performance of the prediction models in the overall validation set and within subgroups defined by race/ethnicity.

The area under the curve measured model discrimination, and sensitivity was estimated for global and race/ethnicity-specific thresholds.
 

‘Unacceptable’ scenario

Within the total population, there were 768 deaths by suicide within 90 days of 3,143 visits. Suicide rates were highest for visits by patients with no recorded race/ethnicity, followed by visits by Asian, White, American Indian/Alaskan Native, Hispanic, and Black patients.

Both models showed “high” AUC sensitivity for White, Hispanic, and Asian patients but “poor” AUC sensitivity for BIPOC and patients without recorded race/ethnicity, the authors reported.

“Implementation of prediction models has to be considered in the broader context of unmet health care needs,” said Dr. Coley.

“In our specific example of suicide prediction, BIPOC populations already face substantial barriers in accessing quality mental health care and, as a result, have poorer outcomes, and using either of the suicide prediction models examined in our study will provide less benefit to already-underserved populations and widen existing care gaps,” a scenario Dr. Coley said is “unacceptable.”

We must insist that new technologies and methods be used to reduce racial and ethnic inequities in care, not exacerbate them,” she added.
 

 

 

Biased algorithms

Commenting on the study, Jonathan Singer, PhD, LCSW, associate professor at Loyola University, Chicago, described it as an “important contribution because it points to a systemic problem and also to the fact that the algorithms we create are biased, created by humans, and humans are biased.”

Although the study focused on the health care system, Dr. Singer believes the findings have implications for individual clinicians.

“If clinicians may be biased against identifying suicide risk in Black and Native American patients, they may attribute suicidal risk to something else. For example, we know that in Black Americans, expressions of intense emotions are oftentimes interpreted as aggression or being threatening, as opposed to indicators of sadness or fear,” noted Dr. Singer, who is also president of the American Academy of Suicidology and was not involved with the study,

“Clinicians who misinterpret these intense emotions are less likely to identify a Black client or patient who is suicidal,” Dr. Singer said.

The research was supported by the Mental Health Research Network from the National Institute of Mental Health. Dr. Coley has reported receiving support through a grant from the Agency for Healthcare Research and Quality. Dr. Singer reported no relevant financial relationships.

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

Current models used to predict suicide risk fall short for racialized populations including Black, Indigenous, and people of color (BIPOC), new research shows.

Investigators developed two suicide prediction models to examine whether these types of tools are accurate in their predictive abilities, or whether they are flawed.

They found both prediction models failed to identify high-risk BIPOC individuals. In the first model, nearly half of outpatient visits followed by suicide were identified in White patients versus only 7% of visits followed by suicide in BIPOC patients. The second model had a sensitivity of 41% for White patients, but just 3% for Black patients and 7% for American Indian/Alaskan Native patients.

Dr. Yates Coley


“You don’t know whether a prediction model will be useful or harmful until it’s evaluated. The take-home message of our study is this: You have to look,” lead author Yates Coley, PhD, assistant investigator, Kaiser Permanente Washington Health Research Institute, Seattle, said in an interview.

The study was published online April 28, 2021, in JAMA Psychiatry.

Racial inequities

Suicide risk prediction models have been “developed and validated in several settings” and are now in regular use at the Veterans Health Administration, HealthPartners, and Kaiser Permanente, the authors wrote.

But the performance of suicide risk prediction models, while accurate in the overall population, “remains unexamined” in particular subpopulations, they noted.

“Health records data reflect existing racial and ethnic inequities in health care access, quality, and outcomes; and prediction models using health records data may perpetuate these disparities by presuming that past healthcare patterns accurately reflect actual needs,” Dr. Coley said.

Dr. Coley and associates “wanted to make sure that any suicide prediction model we implemented in clinical care reduced health disparities rather than exacerbated them.”

To investigate, researchers examined all outpatient mental health visits to seven large integrated health care systems by patients 13 years and older (n = 13,980,570 visits by 1,422,534 patients; 64% female, mean age, 42 years). The study spanned from Jan. 1, 2009, to Sept. 30, 2017, with follow-up through Dec. 31, 2017.

In particular, researchers looked at suicides that took place within 90 days following the outpatient visit.

Researchers used two prediction models: logistic regression with LASSO (Least Absolute Shrinkage and Selection Operator) variable selection and random forest technique, a “tree-based method that explores interactions between predictors (including those with race and ethnicity) in estimating probability of an outcome.”

The models considered prespecified interactions between predictors, including prior diagnoses, suicide attempts, and PHQ-9 [Patient Health Questionnaire–9] responses, and race and ethnicity data.

Researchers evaluated performance of the prediction models in the overall validation set and within subgroups defined by race/ethnicity.

The area under the curve measured model discrimination, and sensitivity was estimated for global and race/ethnicity-specific thresholds.
 

‘Unacceptable’ scenario

Within the total population, there were 768 deaths by suicide within 90 days of 3,143 visits. Suicide rates were highest for visits by patients with no recorded race/ethnicity, followed by visits by Asian, White, American Indian/Alaskan Native, Hispanic, and Black patients.

Both models showed “high” AUC sensitivity for White, Hispanic, and Asian patients but “poor” AUC sensitivity for BIPOC and patients without recorded race/ethnicity, the authors reported.

“Implementation of prediction models has to be considered in the broader context of unmet health care needs,” said Dr. Coley.

“In our specific example of suicide prediction, BIPOC populations already face substantial barriers in accessing quality mental health care and, as a result, have poorer outcomes, and using either of the suicide prediction models examined in our study will provide less benefit to already-underserved populations and widen existing care gaps,” a scenario Dr. Coley said is “unacceptable.”

We must insist that new technologies and methods be used to reduce racial and ethnic inequities in care, not exacerbate them,” she added.
 

 

 

Biased algorithms

Commenting on the study, Jonathan Singer, PhD, LCSW, associate professor at Loyola University, Chicago, described it as an “important contribution because it points to a systemic problem and also to the fact that the algorithms we create are biased, created by humans, and humans are biased.”

Although the study focused on the health care system, Dr. Singer believes the findings have implications for individual clinicians.

“If clinicians may be biased against identifying suicide risk in Black and Native American patients, they may attribute suicidal risk to something else. For example, we know that in Black Americans, expressions of intense emotions are oftentimes interpreted as aggression or being threatening, as opposed to indicators of sadness or fear,” noted Dr. Singer, who is also president of the American Academy of Suicidology and was not involved with the study,

“Clinicians who misinterpret these intense emotions are less likely to identify a Black client or patient who is suicidal,” Dr. Singer said.

The research was supported by the Mental Health Research Network from the National Institute of Mental Health. Dr. Coley has reported receiving support through a grant from the Agency for Healthcare Research and Quality. Dr. Singer reported no relevant financial relationships.

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

FDA authorizes Pfizer COVID vaccine for teens 12-15

Article Type
Changed
Thu, 08/26/2021 - 15:47

 

The Food and Drug Administration on May 10 granted emergency use authorization (EUA) for the Pfizer coronavirus vaccine to be given to children 12-15 years old.  

The much-expected decision increases the likelihood that schools in the United States will fully reopen in the fall – a goal of both the Biden and Trump administrations.

Acting FDA Commissioner Janet Woodcock, MD, called the decision “a significant step” in “returning to a sense of normalcy.”

“Today’s action allows for a younger population to be protected from COVID-19, bringing us closer to returning to a sense of normalcy and to ending the pandemic,” she said in a statement. “Parents and guardians can rest assured that the agency undertook a rigorous and thorough review of all available data, as we have with all of our COVID-19 vaccine emergency use authorizations.”

The Pfizer adolescent vaccine is not yet a done deal, though.

Next, the Centers for Disease Control and Prevention’s Advisory Committee on Immunization Practices will decide on May 12 whether to recommend use of the vaccine in this age group. After that, CDC Director Rochelle Walensky, MD, will decide whether to give the green light for the vaccine to be administered to that age group.

The FDA action on May 10 amends the Dec. 11, 2020, emergency use authorization that allowed the Pfizer vaccine to be given to people 16 and older. Pfizer was the first company to receive an EUA for its adult vaccine and is the first to receive authorization for its adolescent vaccine. Pfizer is conducting clinical trials on much younger children, too.

The Moderna and Johnson & Johnson vaccines are authorized for people 18 and up. Moderna also has launched clinical trials in children.

Most health experts have said the United States needs to vaccinate children before the COVID-19 pandemic can truly be brought under control. The 12- to 15-year-old group represents 17 million people, about 5% of the population. Thus far, 58% of U.S. adults have had at least one dose of a vaccine and 34.8% of all Americans are fully vaccinated.

American Academy of Pediatrics President Lee Savio Beers, MD, praised the agency’s decision, calling it a “critically important step in bringing life-saving vaccines to children and adolescents. Our youngest generations have shouldered heavy burdens over the past year, and the vaccine is a hopeful sign that they will be able to begin to experience all the activities that are so important for their health and development.”

President Joe Biden recently announced a new strategy for expanding vaccinations in which vaccinating 12- to 15-year-olds was a key component. He said the administration was ready to ship the adolescent vaccine directly to pharmacies and pediatricians to speed up the vaccination rate.

In March, Anthony S. Fauci, MD, told a Senate committee, “We don’t really know what that magical point of herd immunity is, but we do know that if we get the overwhelming population vaccinated, we’re going to be in good shape. … We ultimately would like to get and have to get children into that mix.” 

Pfizer submitted data to the FDA in late March showing its mRNA vaccine was 100% effective at preventing COVID-19 infection in children ages 12-15 in clinical trials.

Though most children have milder symptoms when infected with the coronavirus, about 1.5 million cases in children aged 11-17 were reported to the CDC between March 1, 2020, and April 30 of this year, the FDA news release said.

Albert Bourla, CEO of Pfizer, tweeted that “today brings very encouraging news for families and adolescents across the United States.

“While this is a meaningful step forward, we are still in a critical period of combating #COVID19 around the world. In the coming weeks, we hope to continue to receive authorizations from global regulators to support worldwide vaccination efforts,” he said. 

“It’s essential for children to be vaccinated against COVID-19. According to data compiled by the AAP and Children’s Hospital Association, more than 3.8 million children have tested positive for COVID-19 in the United States since the start of the pandemic,” said Dr. Savio Beers. “While fewer children than adults have suffered the most severe disease, this is not a benign disease in children. Thousands of children have been hospitalized, and hundreds have died. We will soon have a very safe, highly effective vaccine that can prevent so much suffering. I encourage parents to talk with their pediatricians about how to get the vaccine for their adolescents as soon as they are eligible.”

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

Publications
Topics
Sections

 

The Food and Drug Administration on May 10 granted emergency use authorization (EUA) for the Pfizer coronavirus vaccine to be given to children 12-15 years old.  

The much-expected decision increases the likelihood that schools in the United States will fully reopen in the fall – a goal of both the Biden and Trump administrations.

Acting FDA Commissioner Janet Woodcock, MD, called the decision “a significant step” in “returning to a sense of normalcy.”

“Today’s action allows for a younger population to be protected from COVID-19, bringing us closer to returning to a sense of normalcy and to ending the pandemic,” she said in a statement. “Parents and guardians can rest assured that the agency undertook a rigorous and thorough review of all available data, as we have with all of our COVID-19 vaccine emergency use authorizations.”

The Pfizer adolescent vaccine is not yet a done deal, though.

Next, the Centers for Disease Control and Prevention’s Advisory Committee on Immunization Practices will decide on May 12 whether to recommend use of the vaccine in this age group. After that, CDC Director Rochelle Walensky, MD, will decide whether to give the green light for the vaccine to be administered to that age group.

The FDA action on May 10 amends the Dec. 11, 2020, emergency use authorization that allowed the Pfizer vaccine to be given to people 16 and older. Pfizer was the first company to receive an EUA for its adult vaccine and is the first to receive authorization for its adolescent vaccine. Pfizer is conducting clinical trials on much younger children, too.

The Moderna and Johnson & Johnson vaccines are authorized for people 18 and up. Moderna also has launched clinical trials in children.

Most health experts have said the United States needs to vaccinate children before the COVID-19 pandemic can truly be brought under control. The 12- to 15-year-old group represents 17 million people, about 5% of the population. Thus far, 58% of U.S. adults have had at least one dose of a vaccine and 34.8% of all Americans are fully vaccinated.

American Academy of Pediatrics President Lee Savio Beers, MD, praised the agency’s decision, calling it a “critically important step in bringing life-saving vaccines to children and adolescents. Our youngest generations have shouldered heavy burdens over the past year, and the vaccine is a hopeful sign that they will be able to begin to experience all the activities that are so important for their health and development.”

President Joe Biden recently announced a new strategy for expanding vaccinations in which vaccinating 12- to 15-year-olds was a key component. He said the administration was ready to ship the adolescent vaccine directly to pharmacies and pediatricians to speed up the vaccination rate.

In March, Anthony S. Fauci, MD, told a Senate committee, “We don’t really know what that magical point of herd immunity is, but we do know that if we get the overwhelming population vaccinated, we’re going to be in good shape. … We ultimately would like to get and have to get children into that mix.” 

Pfizer submitted data to the FDA in late March showing its mRNA vaccine was 100% effective at preventing COVID-19 infection in children ages 12-15 in clinical trials.

Though most children have milder symptoms when infected with the coronavirus, about 1.5 million cases in children aged 11-17 were reported to the CDC between March 1, 2020, and April 30 of this year, the FDA news release said.

Albert Bourla, CEO of Pfizer, tweeted that “today brings very encouraging news for families and adolescents across the United States.

“While this is a meaningful step forward, we are still in a critical period of combating #COVID19 around the world. In the coming weeks, we hope to continue to receive authorizations from global regulators to support worldwide vaccination efforts,” he said. 

“It’s essential for children to be vaccinated against COVID-19. According to data compiled by the AAP and Children’s Hospital Association, more than 3.8 million children have tested positive for COVID-19 in the United States since the start of the pandemic,” said Dr. Savio Beers. “While fewer children than adults have suffered the most severe disease, this is not a benign disease in children. Thousands of children have been hospitalized, and hundreds have died. We will soon have a very safe, highly effective vaccine that can prevent so much suffering. I encourage parents to talk with their pediatricians about how to get the vaccine for their adolescents as soon as they are eligible.”

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

 

The Food and Drug Administration on May 10 granted emergency use authorization (EUA) for the Pfizer coronavirus vaccine to be given to children 12-15 years old.  

The much-expected decision increases the likelihood that schools in the United States will fully reopen in the fall – a goal of both the Biden and Trump administrations.

Acting FDA Commissioner Janet Woodcock, MD, called the decision “a significant step” in “returning to a sense of normalcy.”

“Today’s action allows for a younger population to be protected from COVID-19, bringing us closer to returning to a sense of normalcy and to ending the pandemic,” she said in a statement. “Parents and guardians can rest assured that the agency undertook a rigorous and thorough review of all available data, as we have with all of our COVID-19 vaccine emergency use authorizations.”

The Pfizer adolescent vaccine is not yet a done deal, though.

Next, the Centers for Disease Control and Prevention’s Advisory Committee on Immunization Practices will decide on May 12 whether to recommend use of the vaccine in this age group. After that, CDC Director Rochelle Walensky, MD, will decide whether to give the green light for the vaccine to be administered to that age group.

The FDA action on May 10 amends the Dec. 11, 2020, emergency use authorization that allowed the Pfizer vaccine to be given to people 16 and older. Pfizer was the first company to receive an EUA for its adult vaccine and is the first to receive authorization for its adolescent vaccine. Pfizer is conducting clinical trials on much younger children, too.

The Moderna and Johnson & Johnson vaccines are authorized for people 18 and up. Moderna also has launched clinical trials in children.

Most health experts have said the United States needs to vaccinate children before the COVID-19 pandemic can truly be brought under control. The 12- to 15-year-old group represents 17 million people, about 5% of the population. Thus far, 58% of U.S. adults have had at least one dose of a vaccine and 34.8% of all Americans are fully vaccinated.

American Academy of Pediatrics President Lee Savio Beers, MD, praised the agency’s decision, calling it a “critically important step in bringing life-saving vaccines to children and adolescents. Our youngest generations have shouldered heavy burdens over the past year, and the vaccine is a hopeful sign that they will be able to begin to experience all the activities that are so important for their health and development.”

President Joe Biden recently announced a new strategy for expanding vaccinations in which vaccinating 12- to 15-year-olds was a key component. He said the administration was ready to ship the adolescent vaccine directly to pharmacies and pediatricians to speed up the vaccination rate.

In March, Anthony S. Fauci, MD, told a Senate committee, “We don’t really know what that magical point of herd immunity is, but we do know that if we get the overwhelming population vaccinated, we’re going to be in good shape. … We ultimately would like to get and have to get children into that mix.” 

Pfizer submitted data to the FDA in late March showing its mRNA vaccine was 100% effective at preventing COVID-19 infection in children ages 12-15 in clinical trials.

Though most children have milder symptoms when infected with the coronavirus, about 1.5 million cases in children aged 11-17 were reported to the CDC between March 1, 2020, and April 30 of this year, the FDA news release said.

Albert Bourla, CEO of Pfizer, tweeted that “today brings very encouraging news for families and adolescents across the United States.

“While this is a meaningful step forward, we are still in a critical period of combating #COVID19 around the world. In the coming weeks, we hope to continue to receive authorizations from global regulators to support worldwide vaccination efforts,” he said. 

“It’s essential for children to be vaccinated against COVID-19. According to data compiled by the AAP and Children’s Hospital Association, more than 3.8 million children have tested positive for COVID-19 in the United States since the start of the pandemic,” said Dr. Savio Beers. “While fewer children than adults have suffered the most severe disease, this is not a benign disease in children. Thousands of children have been hospitalized, and hundreds have died. We will soon have a very safe, highly effective vaccine that can prevent so much suffering. I encourage parents to talk with their pediatricians about how to get the vaccine for their adolescents as soon as they are eligible.”

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

Operational changes in primary care linked with improved smoking, blood pressure outcomes

Article Type
Changed
Tue, 05/11/2021 - 13:44

 

Operational changes are linked to improvements in smoking and blood pressure outcomes in primary care practice settings, new research indicates.

The qualitative analysis, published in Annals of Family Medicine , included smoking and blood pressure as separate outcome measures.

The outcomes were calculated using Clinical Quality Measure improvements, with targets of at least 10-point absolute improvements in the proportion of patients with smoking screening, if relevant, counseling, and in the proportion of hypertensive patients with adequately controlled BP. The results were obtained from practices participating in Evidence-NOW, a multisite cardiovascular disease prevention initiative. Configurational Comparative Methods were used to evaluate the joint effects of multiple factors on outcomes.

The majority of practices in the analysis were clinician owned, small (fewer than six clinicians), and/or in an urban location. The researchers sampled and interviewed practice staff from a subset of 104 primary care practices across 7 Cooperatives and 12 states, ranging from small to medium in size, having 10 or fewer clinicians. The interview data were analyzed to identify operational changes, then transformed into numeric data.
 

Operational changes led to improvements in specific contexts

In clinician-owned practices, process improvement, documentation, and referral to resources, combined with a moderate level of facilitation support, led to an improvement of at least 10 points in smoking outcomes.

However, the researchers found that these patterns were not observed in system–owned practices or Federally Qualified Health Centers.

In solo practices, training medical assistants to take an accurate blood pressure led to an improvement of at least 10 points in blood pressure outcomes.

Among larger, clinician-owned practices, measurement of blood pressure a second time when the first was elevated, and documentation of this reading in the electronic heath record, also led to a 10-point or greater improvement in BP outcome when combined with a large amount (50 hours or more) of facilitation.

“There was no magic bullet for improving smoking cessation counseling and blood pressure outcomes across the diverse primary care practices studied,” lead author Deborah J. Cohen, PhD, of Oregon Health & Science University, Portland, said in an interview. “Combinations of operational changes among practice sizes and types led to improvements.”
 

Smaller practices more nimble, experts say

Results of the qualitative data analysis suggest that smaller and clinician-owned practices are more likely to have the capacity for change and improvement compared with larger, hospital/health system–owned practices.

Commenting on the study, Noel Deep, MD, regional medical director at Aspirus Clinics, Ironwood, Mich., said solo or small private practices have a distinct advantage over larger hospital or system-owned practices when implementing new operational changes to improve clinical outcomes.

“A smaller independent practice is nimble, with the physician [or physicians] able to make a quick decision at analyzing the scientific data, planning the changes, implementing them quickly, and doing a rapid cycle review of the results and tweaking the program to attain the targets,” said Dr. Deep, a member of the editorial advisory board of Internal Medicine News.

Kate Rowland, MD, MS, assistant professor in the department of family medicine at Rush Medical College, Chicago, also noted that smaller practices have unique advantages over larger health organizations.

“Larger organizations should replicate the benefits of the smaller office, providing as much local decision-making and autonomy as possible to the site where the changes are happening,” Dr. Rowland explained in an interview.

“The clinicians at these sites are mostly likely to know what is going to be successful for achieving measurable change in the patients they care for,” she added.

The study was funded by the Agency for Healthcare Research and Quality. The authors and other experts interviewed for this piece reported having no conflicts of interest.

Publications
Topics
Sections

 

Operational changes are linked to improvements in smoking and blood pressure outcomes in primary care practice settings, new research indicates.

The qualitative analysis, published in Annals of Family Medicine , included smoking and blood pressure as separate outcome measures.

The outcomes were calculated using Clinical Quality Measure improvements, with targets of at least 10-point absolute improvements in the proportion of patients with smoking screening, if relevant, counseling, and in the proportion of hypertensive patients with adequately controlled BP. The results were obtained from practices participating in Evidence-NOW, a multisite cardiovascular disease prevention initiative. Configurational Comparative Methods were used to evaluate the joint effects of multiple factors on outcomes.

The majority of practices in the analysis were clinician owned, small (fewer than six clinicians), and/or in an urban location. The researchers sampled and interviewed practice staff from a subset of 104 primary care practices across 7 Cooperatives and 12 states, ranging from small to medium in size, having 10 or fewer clinicians. The interview data were analyzed to identify operational changes, then transformed into numeric data.
 

Operational changes led to improvements in specific contexts

In clinician-owned practices, process improvement, documentation, and referral to resources, combined with a moderate level of facilitation support, led to an improvement of at least 10 points in smoking outcomes.

However, the researchers found that these patterns were not observed in system–owned practices or Federally Qualified Health Centers.

In solo practices, training medical assistants to take an accurate blood pressure led to an improvement of at least 10 points in blood pressure outcomes.

Among larger, clinician-owned practices, measurement of blood pressure a second time when the first was elevated, and documentation of this reading in the electronic heath record, also led to a 10-point or greater improvement in BP outcome when combined with a large amount (50 hours or more) of facilitation.

“There was no magic bullet for improving smoking cessation counseling and blood pressure outcomes across the diverse primary care practices studied,” lead author Deborah J. Cohen, PhD, of Oregon Health & Science University, Portland, said in an interview. “Combinations of operational changes among practice sizes and types led to improvements.”
 

Smaller practices more nimble, experts say

Results of the qualitative data analysis suggest that smaller and clinician-owned practices are more likely to have the capacity for change and improvement compared with larger, hospital/health system–owned practices.

Commenting on the study, Noel Deep, MD, regional medical director at Aspirus Clinics, Ironwood, Mich., said solo or small private practices have a distinct advantage over larger hospital or system-owned practices when implementing new operational changes to improve clinical outcomes.

“A smaller independent practice is nimble, with the physician [or physicians] able to make a quick decision at analyzing the scientific data, planning the changes, implementing them quickly, and doing a rapid cycle review of the results and tweaking the program to attain the targets,” said Dr. Deep, a member of the editorial advisory board of Internal Medicine News.

Kate Rowland, MD, MS, assistant professor in the department of family medicine at Rush Medical College, Chicago, also noted that smaller practices have unique advantages over larger health organizations.

“Larger organizations should replicate the benefits of the smaller office, providing as much local decision-making and autonomy as possible to the site where the changes are happening,” Dr. Rowland explained in an interview.

“The clinicians at these sites are mostly likely to know what is going to be successful for achieving measurable change in the patients they care for,” she added.

The study was funded by the Agency for Healthcare Research and Quality. The authors and other experts interviewed for this piece reported having no conflicts of interest.

 

Operational changes are linked to improvements in smoking and blood pressure outcomes in primary care practice settings, new research indicates.

The qualitative analysis, published in Annals of Family Medicine , included smoking and blood pressure as separate outcome measures.

The outcomes were calculated using Clinical Quality Measure improvements, with targets of at least 10-point absolute improvements in the proportion of patients with smoking screening, if relevant, counseling, and in the proportion of hypertensive patients with adequately controlled BP. The results were obtained from practices participating in Evidence-NOW, a multisite cardiovascular disease prevention initiative. Configurational Comparative Methods were used to evaluate the joint effects of multiple factors on outcomes.

The majority of practices in the analysis were clinician owned, small (fewer than six clinicians), and/or in an urban location. The researchers sampled and interviewed practice staff from a subset of 104 primary care practices across 7 Cooperatives and 12 states, ranging from small to medium in size, having 10 or fewer clinicians. The interview data were analyzed to identify operational changes, then transformed into numeric data.
 

Operational changes led to improvements in specific contexts

In clinician-owned practices, process improvement, documentation, and referral to resources, combined with a moderate level of facilitation support, led to an improvement of at least 10 points in smoking outcomes.

However, the researchers found that these patterns were not observed in system–owned practices or Federally Qualified Health Centers.

In solo practices, training medical assistants to take an accurate blood pressure led to an improvement of at least 10 points in blood pressure outcomes.

Among larger, clinician-owned practices, measurement of blood pressure a second time when the first was elevated, and documentation of this reading in the electronic heath record, also led to a 10-point or greater improvement in BP outcome when combined with a large amount (50 hours or more) of facilitation.

“There was no magic bullet for improving smoking cessation counseling and blood pressure outcomes across the diverse primary care practices studied,” lead author Deborah J. Cohen, PhD, of Oregon Health & Science University, Portland, said in an interview. “Combinations of operational changes among practice sizes and types led to improvements.”
 

Smaller practices more nimble, experts say

Results of the qualitative data analysis suggest that smaller and clinician-owned practices are more likely to have the capacity for change and improvement compared with larger, hospital/health system–owned practices.

Commenting on the study, Noel Deep, MD, regional medical director at Aspirus Clinics, Ironwood, Mich., said solo or small private practices have a distinct advantage over larger hospital or system-owned practices when implementing new operational changes to improve clinical outcomes.

“A smaller independent practice is nimble, with the physician [or physicians] able to make a quick decision at analyzing the scientific data, planning the changes, implementing them quickly, and doing a rapid cycle review of the results and tweaking the program to attain the targets,” said Dr. Deep, a member of the editorial advisory board of Internal Medicine News.

Kate Rowland, MD, MS, assistant professor in the department of family medicine at Rush Medical College, Chicago, also noted that smaller practices have unique advantages over larger health organizations.

“Larger organizations should replicate the benefits of the smaller office, providing as much local decision-making and autonomy as possible to the site where the changes are happening,” Dr. Rowland explained in an interview.

“The clinicians at these sites are mostly likely to know what is going to be successful for achieving measurable change in the patients they care for,” she added.

The study was funded by the Agency for Healthcare Research and Quality. The authors and other experts interviewed for this piece reported having no conflicts of interest.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM ANNALS OF FAMILY MEDICINE

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

Low-fat diet upped quality of life in ulcerative colitis

Much-needed diet science sought
Article Type
Changed
Tue, 05/11/2021 - 11:41

For patients with mild or remitted ulcerative colitis, a catered, low-fat, high-fiber diet improved quality of life and stool markers of dysbiosis and inflammation, according to the findings of a small crossover trial.

Lisovskaya/iStock/Getty Images

Patients with inflammatory bowel disease often ask what they should eat, but few studies have addressed that question, Julia Fritsch, of the University of Miami and her associates wrote in Clinical Gastroenterology and Hepatology. Building on previous findings that a high-fat diet may contribute to inflammatory bowel disease, they randomly assigned 38 adults whose ulcerative colitis was in remission or mild (with a flare within the past 18 months) to receive either a low-fat diet (with 10% of daily calories from fat and high amounts of fruit and vegetables) or an “improved American standard diet” (with 35%-40% of daily calories from fat but more fruit and vegetables than Americans typically eat). Each diet was catered, delivered to patients’ homes, and lasted 4 weeks, followed by a 2-week washout period, after which each participant switched to the other diet.

Of the 38 patients, 17 completed the study. Food recall surveys over 24 hours showed that both diets were healthier than what participants ate at baseline, and daily web-based food diaries (such as www.nutrihand.com/Static/index.html) confirmed that more than 94% of patients adhered to the amount of fat in each diet. Even though participants in both groups ate only about half of the provided fruits and vegetables, the primary outcome of quality of life based on the short inflammatory bowel disease questionnaire (SIBDQ) significantly improved from a median of 4.98 (interquartile range, 4.1-6.0) at baseline to 5.77 (IQR, 5-6.4) with the low-fat diet and 5.55 (IQR, 4.75-6.25) with the improved American standard diet. Both diets also produced significant improvements in quality of life as measured by the 36-Item Short Form Survey and in disease activity as measured by the partial Mayo score.

Notably, however, only the low-fat diet significantly reduced serum amyloid A, which is a marker of mucosal inflammation, and intestinal dysbiosis, which was quantified by 16S RNA ribosomal sequencing. “Of note, there were several variables that were associated with changes in the microbiota composition,” the researchers wrote. These included the SIBDQ, C-reactive protein, interleukin-6, interleukin-1 beta, and 32 dietary components such as protein, potassium, iron, and zinc.

“These data suggest that even patients in remission [from ulcerative colitis] could benefit from a healthier diet,” the investigators concluded. “Just as importantly, neither diet exacerbated symptoms, which is notable given the higher fiber in both catered diets.” They called catering “a feasible way to perform a diet intervention study with high adherence,” noting that “catering a diet for a patient with IBD for a year costs between $19,000 and $21,000 per patient. The cost of a patient on a biologic such as ustekinumab is approximately $130,752 to $261,504.”

The study was supported by the Crohn’s and Colitis Foundation Broad Medical Research Program, Micky and Madeleine Arison Family Foundation Crohn’s and Colitis Discovery Laboratory, and the Martin Kalser Chair. The senior author disclosed ties to Boehringer Ingelheim, Gilead, AbbVie, Seres Therapeutics, Shire, Landos, Pfizer, and several other pharmaceutical companies. The other researchers reported having no conflicts of interest.

Body

 

Diet plays an important role in Crohn’s disease and ulcerative colitis. Most patients with these diseases look to incorporate dietary modification as part of the treatment plan to achieve and maintain remission. With the development of tools that allow us to sequence the gut microbiome at high resolution, the role of dietary therapy for these diseases is being studied with increasing scientific rigor.

In a crossover study of 17 patients with ulcerative colitis in remission or with only mild disease, Fritsch and colleagues demonstrated that adherence to a low-fat, high-fiber diet was associated with an improvement in the health-related quality of life, a decrease in C-reactive protein, and beneficial changes in the gut bacteria including reduced abundance of Actinobacteria and an increase in organisms with anti-inflammatory potential such as Faecalibacterium prausnitzii. In conjunction with prior experimental studies that suggested an increase in risk of colitis with high fat intake, this study provides some evidence for recommending a lower fat intake in patients with established inflammatory bowel disease (IBD). Furthermore, an increase in fruits, vegetables and fiber intake even in those with a standard American diet was associated with a modest beneficial effect, challenging the longstanding unsupported dogma of broadly limiting all fiber intake in those with established IBD.

The much-needed progress in the scientific study of diet in IBD will provide us with the important answers that our patients are looking for.
 

Ashwin Ananthakrishnan, MD, MPH , is an associate professor of medicine at Massachusetts General Hospital and Harvard Medical School, both in Boston. He has no conflicts relevant to this commentary to declare.

Publications
Topics
Sections
Body

 

Diet plays an important role in Crohn’s disease and ulcerative colitis. Most patients with these diseases look to incorporate dietary modification as part of the treatment plan to achieve and maintain remission. With the development of tools that allow us to sequence the gut microbiome at high resolution, the role of dietary therapy for these diseases is being studied with increasing scientific rigor.

In a crossover study of 17 patients with ulcerative colitis in remission or with only mild disease, Fritsch and colleagues demonstrated that adherence to a low-fat, high-fiber diet was associated with an improvement in the health-related quality of life, a decrease in C-reactive protein, and beneficial changes in the gut bacteria including reduced abundance of Actinobacteria and an increase in organisms with anti-inflammatory potential such as Faecalibacterium prausnitzii. In conjunction with prior experimental studies that suggested an increase in risk of colitis with high fat intake, this study provides some evidence for recommending a lower fat intake in patients with established inflammatory bowel disease (IBD). Furthermore, an increase in fruits, vegetables and fiber intake even in those with a standard American diet was associated with a modest beneficial effect, challenging the longstanding unsupported dogma of broadly limiting all fiber intake in those with established IBD.

The much-needed progress in the scientific study of diet in IBD will provide us with the important answers that our patients are looking for.
 

Ashwin Ananthakrishnan, MD, MPH , is an associate professor of medicine at Massachusetts General Hospital and Harvard Medical School, both in Boston. He has no conflicts relevant to this commentary to declare.

Body

 

Diet plays an important role in Crohn’s disease and ulcerative colitis. Most patients with these diseases look to incorporate dietary modification as part of the treatment plan to achieve and maintain remission. With the development of tools that allow us to sequence the gut microbiome at high resolution, the role of dietary therapy for these diseases is being studied with increasing scientific rigor.

In a crossover study of 17 patients with ulcerative colitis in remission or with only mild disease, Fritsch and colleagues demonstrated that adherence to a low-fat, high-fiber diet was associated with an improvement in the health-related quality of life, a decrease in C-reactive protein, and beneficial changes in the gut bacteria including reduced abundance of Actinobacteria and an increase in organisms with anti-inflammatory potential such as Faecalibacterium prausnitzii. In conjunction with prior experimental studies that suggested an increase in risk of colitis with high fat intake, this study provides some evidence for recommending a lower fat intake in patients with established inflammatory bowel disease (IBD). Furthermore, an increase in fruits, vegetables and fiber intake even in those with a standard American diet was associated with a modest beneficial effect, challenging the longstanding unsupported dogma of broadly limiting all fiber intake in those with established IBD.

The much-needed progress in the scientific study of diet in IBD will provide us with the important answers that our patients are looking for.
 

Ashwin Ananthakrishnan, MD, MPH , is an associate professor of medicine at Massachusetts General Hospital and Harvard Medical School, both in Boston. He has no conflicts relevant to this commentary to declare.

Title
Much-needed diet science sought
Much-needed diet science sought

For patients with mild or remitted ulcerative colitis, a catered, low-fat, high-fiber diet improved quality of life and stool markers of dysbiosis and inflammation, according to the findings of a small crossover trial.

Lisovskaya/iStock/Getty Images

Patients with inflammatory bowel disease often ask what they should eat, but few studies have addressed that question, Julia Fritsch, of the University of Miami and her associates wrote in Clinical Gastroenterology and Hepatology. Building on previous findings that a high-fat diet may contribute to inflammatory bowel disease, they randomly assigned 38 adults whose ulcerative colitis was in remission or mild (with a flare within the past 18 months) to receive either a low-fat diet (with 10% of daily calories from fat and high amounts of fruit and vegetables) or an “improved American standard diet” (with 35%-40% of daily calories from fat but more fruit and vegetables than Americans typically eat). Each diet was catered, delivered to patients’ homes, and lasted 4 weeks, followed by a 2-week washout period, after which each participant switched to the other diet.

Of the 38 patients, 17 completed the study. Food recall surveys over 24 hours showed that both diets were healthier than what participants ate at baseline, and daily web-based food diaries (such as www.nutrihand.com/Static/index.html) confirmed that more than 94% of patients adhered to the amount of fat in each diet. Even though participants in both groups ate only about half of the provided fruits and vegetables, the primary outcome of quality of life based on the short inflammatory bowel disease questionnaire (SIBDQ) significantly improved from a median of 4.98 (interquartile range, 4.1-6.0) at baseline to 5.77 (IQR, 5-6.4) with the low-fat diet and 5.55 (IQR, 4.75-6.25) with the improved American standard diet. Both diets also produced significant improvements in quality of life as measured by the 36-Item Short Form Survey and in disease activity as measured by the partial Mayo score.

Notably, however, only the low-fat diet significantly reduced serum amyloid A, which is a marker of mucosal inflammation, and intestinal dysbiosis, which was quantified by 16S RNA ribosomal sequencing. “Of note, there were several variables that were associated with changes in the microbiota composition,” the researchers wrote. These included the SIBDQ, C-reactive protein, interleukin-6, interleukin-1 beta, and 32 dietary components such as protein, potassium, iron, and zinc.

“These data suggest that even patients in remission [from ulcerative colitis] could benefit from a healthier diet,” the investigators concluded. “Just as importantly, neither diet exacerbated symptoms, which is notable given the higher fiber in both catered diets.” They called catering “a feasible way to perform a diet intervention study with high adherence,” noting that “catering a diet for a patient with IBD for a year costs between $19,000 and $21,000 per patient. The cost of a patient on a biologic such as ustekinumab is approximately $130,752 to $261,504.”

The study was supported by the Crohn’s and Colitis Foundation Broad Medical Research Program, Micky and Madeleine Arison Family Foundation Crohn’s and Colitis Discovery Laboratory, and the Martin Kalser Chair. The senior author disclosed ties to Boehringer Ingelheim, Gilead, AbbVie, Seres Therapeutics, Shire, Landos, Pfizer, and several other pharmaceutical companies. The other researchers reported having no conflicts of interest.

For patients with mild or remitted ulcerative colitis, a catered, low-fat, high-fiber diet improved quality of life and stool markers of dysbiosis and inflammation, according to the findings of a small crossover trial.

Lisovskaya/iStock/Getty Images

Patients with inflammatory bowel disease often ask what they should eat, but few studies have addressed that question, Julia Fritsch, of the University of Miami and her associates wrote in Clinical Gastroenterology and Hepatology. Building on previous findings that a high-fat diet may contribute to inflammatory bowel disease, they randomly assigned 38 adults whose ulcerative colitis was in remission or mild (with a flare within the past 18 months) to receive either a low-fat diet (with 10% of daily calories from fat and high amounts of fruit and vegetables) or an “improved American standard diet” (with 35%-40% of daily calories from fat but more fruit and vegetables than Americans typically eat). Each diet was catered, delivered to patients’ homes, and lasted 4 weeks, followed by a 2-week washout period, after which each participant switched to the other diet.

Of the 38 patients, 17 completed the study. Food recall surveys over 24 hours showed that both diets were healthier than what participants ate at baseline, and daily web-based food diaries (such as www.nutrihand.com/Static/index.html) confirmed that more than 94% of patients adhered to the amount of fat in each diet. Even though participants in both groups ate only about half of the provided fruits and vegetables, the primary outcome of quality of life based on the short inflammatory bowel disease questionnaire (SIBDQ) significantly improved from a median of 4.98 (interquartile range, 4.1-6.0) at baseline to 5.77 (IQR, 5-6.4) with the low-fat diet and 5.55 (IQR, 4.75-6.25) with the improved American standard diet. Both diets also produced significant improvements in quality of life as measured by the 36-Item Short Form Survey and in disease activity as measured by the partial Mayo score.

Notably, however, only the low-fat diet significantly reduced serum amyloid A, which is a marker of mucosal inflammation, and intestinal dysbiosis, which was quantified by 16S RNA ribosomal sequencing. “Of note, there were several variables that were associated with changes in the microbiota composition,” the researchers wrote. These included the SIBDQ, C-reactive protein, interleukin-6, interleukin-1 beta, and 32 dietary components such as protein, potassium, iron, and zinc.

“These data suggest that even patients in remission [from ulcerative colitis] could benefit from a healthier diet,” the investigators concluded. “Just as importantly, neither diet exacerbated symptoms, which is notable given the higher fiber in both catered diets.” They called catering “a feasible way to perform a diet intervention study with high adherence,” noting that “catering a diet for a patient with IBD for a year costs between $19,000 and $21,000 per patient. The cost of a patient on a biologic such as ustekinumab is approximately $130,752 to $261,504.”

The study was supported by the Crohn’s and Colitis Foundation Broad Medical Research Program, Micky and Madeleine Arison Family Foundation Crohn’s and Colitis Discovery Laboratory, and the Martin Kalser Chair. The senior author disclosed ties to Boehringer Ingelheim, Gilead, AbbVie, Seres Therapeutics, Shire, Landos, Pfizer, and several other pharmaceutical companies. The other researchers reported having no conflicts of interest.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY

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

IL-6 trans-signaling targeted by olamkicept in IBD

Science perseveres – and eventually succeeds?
Article Type
Changed
Tue, 05/18/2021 - 13:06

 

The selective interleukin-6 (IL-6) trans-signaling inhibitor olamkicept was well tolerated and induced clinical remissions in 3 of 16 adults with moderately to severely active inflammatory bowel disease (IBD), and remission was associated with clear alterations in levels of phospho-STAT3 (pSTAT3) in the intestinal mucosa, researchers reported.

In a 12-week, open-label, prospective phase 2a trial, patients received up to seven infusions of 600-mg olamkicept (sgp130Fc) every 2 weeks. Clinical remissions occurred in two of nine patients with ulcerative colitis and one of seven patients with Crohn’s disease. The overall rate of clinical response was 44%, which included five patients with ulcerative colitis and two patients with Crohn’s disease. Transcriptome isolation and high-throughput RNA sequencing of mucosal tissue specimens showed that clinical remitters had a decrease from baseline to week 14 in the expression of TNF, IL-1A, REG1A, IL-8, IL-1B, and LILRA, a known composite molecular surrogate for mucosal inflammation. In addition, exposing whole-blood samples to a recombinant IL-6/IL-6R fusion protein mimicked physiologic IL-6 activity and demonstrated that pSTAT3 levels dropped within 4 hours of the first olamkicept infusion and throughout treatment. “Our overall finding of decreased pSTAT3-positive cells in remission patients indicates that STAT3 is crucially involved in the mechanism of action of olamkicept,” wrote Stefan Schreiber, MD, of University Medical Center Schleswig-Holstein, Campus Kiel (Germany) together with his associates. The study is published in Gastroenterology.

Blocking the IL-6/ILR receptor can induce IBD remissions but causes “profound immunosuppression,” the investigators noted. Building on prior findings that chronic proinflammatory IL-6 activity is primarily mediated by trans-signaling of a complex of IL-6 and soluble IL6R that engages the gp130 receptor, the researchers developed a “decoy protein,” sgp130Fc (now known as olamkicept), which “exclusively blocks” IL-6 proinflammatory trans-signaling. This decoy protein showed promise in preclinical studies, with no evidence of immunosuppression, they wrote. To further evaluate olamkicept, they recruited adults with moderately to severely active ulcerative colitis or Crohn’s disease from two centers in Germany. The primary clinical assessment was remission, defined as a Mayo score under 2, with a bleeding score of 0 and an endoscopy score of less than 1 for patients with ulcerative colitis, and a Crohn’s Disease Activity Index (CDAI) of less than 150 for patients with Crohn’s disease. The primary molecular outcome was change in the composite molecular surrogate score.

Of the 16 patients, 10 completed the trial. At week 14, endoscopic responses were observed in six patients, all of whom also had a clinical response, and all three patients with clinical remissions also had endoscopic remissions. “The drug was well tolerated in all 16 treated individuals, similar to the results of the [two prior] phase 1 trials,” the researchers wrote. Although significant immunosuppression and intestinal perforations were not seen, 13 patients developed adverse events, most commonly seasonal upper respiratory tract infections, recurrence of herpes labialis, and eczema or erythema. There were five serious adverse events, two of which were cardiac in nature. A larger placebo-controlled trial is underway to further evaluate safety. For now, the researchers wrote, it appears that IL-6 trans-signaling inhibition “might open up novel therapeutic avenues for the treatment of IBD.”

University Hospital Schleswig-Holstein sponsored the study. Ferring AG provided funding and donated the olamkicept. Analyses were funded by EU H2020 SYSCID and EU H2020 Innovative Medicines Initiative 2 Joint Undertaking. Dr. Schreiber reported having coinvented IP and having ties to Pfizer, Bristol Myers Squibb, and Roche. Four coinvestigators disclosed ties to Ferring, AbbVie, Chugai, Roche, Regeneron, Pfizer, Sanofi, Conaris, and Genentech Roche. The other researchers reported having no conflicts of interest.

Body

 

Proinflammatory cytokine inhibition has revolutionized the care of patients with moderate to severe inflammatory bowel disease (IBD). However, some patients don’t respond, never gain remission, or lose response. Therefore, the search continues for more effective therapies. The study by Schreiber and colleagues highlights the importance of continued innovation surrounding inflammatory pathways.

Vanderbilt University
Dr. Sara Horst
In the early 2000s, clinical trials were undertaken with an IL-6R monoclonal antibody in Crohn’s disease. These trials showed efficacy, but patients had significant serious adverse events secondary to excessive immunosuppression including abscesses, perforation, and death. Encouragingly, several of the patients with IBD in this small phase 2a, 12-week, open-label trial showed a clinical response. 

The authors did extensive evaluation of the tissue and molecular effects and discovered possible differential target engagement with interleukin-6 transcriptional inhibition which is encouraging. Notably, however, there were a high number of reported adverse events. Per the authors, these were nonspecific and not indicative of severe immunosuppression. Importantly, there were no intestinal perforations.

Intense optimism for new mechanisms will remain tempered as we have seen other therapies hold promise but fail in larger randomized trials. However, it is encouraging to see how continued work on proinflammatory pathways into more targeted inhibitory approaches can lead to potential new therapies in IBD.

Sara Horst, MD, MPH, FACG, is an associate professor in the division of gastroenterology, hepatology, and nutrition at Vanderbilt University Medical Center, Nashville, Tenn. She reports having been a consultant for Gilead, Takeda, and Janssen and receiving unrestricted grant funding from UCB.

Publications
Topics
Sections
Body

 

Proinflammatory cytokine inhibition has revolutionized the care of patients with moderate to severe inflammatory bowel disease (IBD). However, some patients don’t respond, never gain remission, or lose response. Therefore, the search continues for more effective therapies. The study by Schreiber and colleagues highlights the importance of continued innovation surrounding inflammatory pathways.

Vanderbilt University
Dr. Sara Horst
In the early 2000s, clinical trials were undertaken with an IL-6R monoclonal antibody in Crohn’s disease. These trials showed efficacy, but patients had significant serious adverse events secondary to excessive immunosuppression including abscesses, perforation, and death. Encouragingly, several of the patients with IBD in this small phase 2a, 12-week, open-label trial showed a clinical response. 

The authors did extensive evaluation of the tissue and molecular effects and discovered possible differential target engagement with interleukin-6 transcriptional inhibition which is encouraging. Notably, however, there were a high number of reported adverse events. Per the authors, these were nonspecific and not indicative of severe immunosuppression. Importantly, there were no intestinal perforations.

Intense optimism for new mechanisms will remain tempered as we have seen other therapies hold promise but fail in larger randomized trials. However, it is encouraging to see how continued work on proinflammatory pathways into more targeted inhibitory approaches can lead to potential new therapies in IBD.

Sara Horst, MD, MPH, FACG, is an associate professor in the division of gastroenterology, hepatology, and nutrition at Vanderbilt University Medical Center, Nashville, Tenn. She reports having been a consultant for Gilead, Takeda, and Janssen and receiving unrestricted grant funding from UCB.

Body

 

Proinflammatory cytokine inhibition has revolutionized the care of patients with moderate to severe inflammatory bowel disease (IBD). However, some patients don’t respond, never gain remission, or lose response. Therefore, the search continues for more effective therapies. The study by Schreiber and colleagues highlights the importance of continued innovation surrounding inflammatory pathways.

Vanderbilt University
Dr. Sara Horst
In the early 2000s, clinical trials were undertaken with an IL-6R monoclonal antibody in Crohn’s disease. These trials showed efficacy, but patients had significant serious adverse events secondary to excessive immunosuppression including abscesses, perforation, and death. Encouragingly, several of the patients with IBD in this small phase 2a, 12-week, open-label trial showed a clinical response. 

The authors did extensive evaluation of the tissue and molecular effects and discovered possible differential target engagement with interleukin-6 transcriptional inhibition which is encouraging. Notably, however, there were a high number of reported adverse events. Per the authors, these were nonspecific and not indicative of severe immunosuppression. Importantly, there were no intestinal perforations.

Intense optimism for new mechanisms will remain tempered as we have seen other therapies hold promise but fail in larger randomized trials. However, it is encouraging to see how continued work on proinflammatory pathways into more targeted inhibitory approaches can lead to potential new therapies in IBD.

Sara Horst, MD, MPH, FACG, is an associate professor in the division of gastroenterology, hepatology, and nutrition at Vanderbilt University Medical Center, Nashville, Tenn. She reports having been a consultant for Gilead, Takeda, and Janssen and receiving unrestricted grant funding from UCB.

Title
Science perseveres – and eventually succeeds?
Science perseveres – and eventually succeeds?

 

The selective interleukin-6 (IL-6) trans-signaling inhibitor olamkicept was well tolerated and induced clinical remissions in 3 of 16 adults with moderately to severely active inflammatory bowel disease (IBD), and remission was associated with clear alterations in levels of phospho-STAT3 (pSTAT3) in the intestinal mucosa, researchers reported.

In a 12-week, open-label, prospective phase 2a trial, patients received up to seven infusions of 600-mg olamkicept (sgp130Fc) every 2 weeks. Clinical remissions occurred in two of nine patients with ulcerative colitis and one of seven patients with Crohn’s disease. The overall rate of clinical response was 44%, which included five patients with ulcerative colitis and two patients with Crohn’s disease. Transcriptome isolation and high-throughput RNA sequencing of mucosal tissue specimens showed that clinical remitters had a decrease from baseline to week 14 in the expression of TNF, IL-1A, REG1A, IL-8, IL-1B, and LILRA, a known composite molecular surrogate for mucosal inflammation. In addition, exposing whole-blood samples to a recombinant IL-6/IL-6R fusion protein mimicked physiologic IL-6 activity and demonstrated that pSTAT3 levels dropped within 4 hours of the first olamkicept infusion and throughout treatment. “Our overall finding of decreased pSTAT3-positive cells in remission patients indicates that STAT3 is crucially involved in the mechanism of action of olamkicept,” wrote Stefan Schreiber, MD, of University Medical Center Schleswig-Holstein, Campus Kiel (Germany) together with his associates. The study is published in Gastroenterology.

Blocking the IL-6/ILR receptor can induce IBD remissions but causes “profound immunosuppression,” the investigators noted. Building on prior findings that chronic proinflammatory IL-6 activity is primarily mediated by trans-signaling of a complex of IL-6 and soluble IL6R that engages the gp130 receptor, the researchers developed a “decoy protein,” sgp130Fc (now known as olamkicept), which “exclusively blocks” IL-6 proinflammatory trans-signaling. This decoy protein showed promise in preclinical studies, with no evidence of immunosuppression, they wrote. To further evaluate olamkicept, they recruited adults with moderately to severely active ulcerative colitis or Crohn’s disease from two centers in Germany. The primary clinical assessment was remission, defined as a Mayo score under 2, with a bleeding score of 0 and an endoscopy score of less than 1 for patients with ulcerative colitis, and a Crohn’s Disease Activity Index (CDAI) of less than 150 for patients with Crohn’s disease. The primary molecular outcome was change in the composite molecular surrogate score.

Of the 16 patients, 10 completed the trial. At week 14, endoscopic responses were observed in six patients, all of whom also had a clinical response, and all three patients with clinical remissions also had endoscopic remissions. “The drug was well tolerated in all 16 treated individuals, similar to the results of the [two prior] phase 1 trials,” the researchers wrote. Although significant immunosuppression and intestinal perforations were not seen, 13 patients developed adverse events, most commonly seasonal upper respiratory tract infections, recurrence of herpes labialis, and eczema or erythema. There were five serious adverse events, two of which were cardiac in nature. A larger placebo-controlled trial is underway to further evaluate safety. For now, the researchers wrote, it appears that IL-6 trans-signaling inhibition “might open up novel therapeutic avenues for the treatment of IBD.”

University Hospital Schleswig-Holstein sponsored the study. Ferring AG provided funding and donated the olamkicept. Analyses were funded by EU H2020 SYSCID and EU H2020 Innovative Medicines Initiative 2 Joint Undertaking. Dr. Schreiber reported having coinvented IP and having ties to Pfizer, Bristol Myers Squibb, and Roche. Four coinvestigators disclosed ties to Ferring, AbbVie, Chugai, Roche, Regeneron, Pfizer, Sanofi, Conaris, and Genentech Roche. The other researchers reported having no conflicts of interest.

 

The selective interleukin-6 (IL-6) trans-signaling inhibitor olamkicept was well tolerated and induced clinical remissions in 3 of 16 adults with moderately to severely active inflammatory bowel disease (IBD), and remission was associated with clear alterations in levels of phospho-STAT3 (pSTAT3) in the intestinal mucosa, researchers reported.

In a 12-week, open-label, prospective phase 2a trial, patients received up to seven infusions of 600-mg olamkicept (sgp130Fc) every 2 weeks. Clinical remissions occurred in two of nine patients with ulcerative colitis and one of seven patients with Crohn’s disease. The overall rate of clinical response was 44%, which included five patients with ulcerative colitis and two patients with Crohn’s disease. Transcriptome isolation and high-throughput RNA sequencing of mucosal tissue specimens showed that clinical remitters had a decrease from baseline to week 14 in the expression of TNF, IL-1A, REG1A, IL-8, IL-1B, and LILRA, a known composite molecular surrogate for mucosal inflammation. In addition, exposing whole-blood samples to a recombinant IL-6/IL-6R fusion protein mimicked physiologic IL-6 activity and demonstrated that pSTAT3 levels dropped within 4 hours of the first olamkicept infusion and throughout treatment. “Our overall finding of decreased pSTAT3-positive cells in remission patients indicates that STAT3 is crucially involved in the mechanism of action of olamkicept,” wrote Stefan Schreiber, MD, of University Medical Center Schleswig-Holstein, Campus Kiel (Germany) together with his associates. The study is published in Gastroenterology.

Blocking the IL-6/ILR receptor can induce IBD remissions but causes “profound immunosuppression,” the investigators noted. Building on prior findings that chronic proinflammatory IL-6 activity is primarily mediated by trans-signaling of a complex of IL-6 and soluble IL6R that engages the gp130 receptor, the researchers developed a “decoy protein,” sgp130Fc (now known as olamkicept), which “exclusively blocks” IL-6 proinflammatory trans-signaling. This decoy protein showed promise in preclinical studies, with no evidence of immunosuppression, they wrote. To further evaluate olamkicept, they recruited adults with moderately to severely active ulcerative colitis or Crohn’s disease from two centers in Germany. The primary clinical assessment was remission, defined as a Mayo score under 2, with a bleeding score of 0 and an endoscopy score of less than 1 for patients with ulcerative colitis, and a Crohn’s Disease Activity Index (CDAI) of less than 150 for patients with Crohn’s disease. The primary molecular outcome was change in the composite molecular surrogate score.

Of the 16 patients, 10 completed the trial. At week 14, endoscopic responses were observed in six patients, all of whom also had a clinical response, and all three patients with clinical remissions also had endoscopic remissions. “The drug was well tolerated in all 16 treated individuals, similar to the results of the [two prior] phase 1 trials,” the researchers wrote. Although significant immunosuppression and intestinal perforations were not seen, 13 patients developed adverse events, most commonly seasonal upper respiratory tract infections, recurrence of herpes labialis, and eczema or erythema. There were five serious adverse events, two of which were cardiac in nature. A larger placebo-controlled trial is underway to further evaluate safety. For now, the researchers wrote, it appears that IL-6 trans-signaling inhibition “might open up novel therapeutic avenues for the treatment of IBD.”

University Hospital Schleswig-Holstein sponsored the study. Ferring AG provided funding and donated the olamkicept. Analyses were funded by EU H2020 SYSCID and EU H2020 Innovative Medicines Initiative 2 Joint Undertaking. Dr. Schreiber reported having coinvented IP and having ties to Pfizer, Bristol Myers Squibb, and Roche. Four coinvestigators disclosed ties to Ferring, AbbVie, Chugai, Roche, Regeneron, Pfizer, Sanofi, Conaris, and Genentech Roche. The other researchers reported having no conflicts of interest.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM GASTROENTEROLOGY

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