Androgen deprivation therapy linked to depression

Article Type
Changed
Fri, 01/04/2019 - 13:14
Display Headline
Androgen deprivation therapy linked to depression

Men on androgen deprivation therapy for prostate cancer are at significantly increased risk for depression, a risk that increases with duration of therapy, investigators report.

A review of Surveillance, Epidemiology, and End Results (SEER) Medicare data on nearly 79,000 men older than 65 years with a diagnosis of prostate cancer showed that those who received androgen deprivation therapy (ADT) had a 23% increased risk for depression, compared with men who were not on ADT, reported Kathryn T. Dinh of Harvard Medical School, Boston, and her colleagues.

“We observed a significantly increased risk of depression and inpatient psychiatric treatment in men treated with ADT for prostate cancer, as well as a duration-response effect such that more ADT was linked to an increasing risk of depression and inpatient and outpatient psychiatric treatment. The possible psychiatric effects of ADT should be recognized by physicians and discussed with patients before initiating treatment,” they wrote (J Clin Oncol. 2016 Apr 11. doi: 10.1200/JCO.2015.64.1969).

 

Although ADT has been identified in some studies as a risk factor for clinical depression, evidence for such a relationship has been spotty, the investigators said, prompting them to conduct a population-based retrospective study to get a better handle on the issue.

They reviewed SEER Medicare data on 78,552 men older than 65 years with a diagnosis of stage I-III prostate cancer treated with ADT from 1992 through 2006, excluding from the sample those patients who had a psychiatric diagnosis within the past 12 months.

Ms. Dinh and her associates found that the 33,882 patients (43%) who received ADT had a significantly higher 3-year cumulative incidence of depression than patients who did not have ADT (7.1% vs. 5.2%, P less than .001), and a significantly higher proportion had either inpatient psychiatric treatment (2.8% vs. 1.9%, P less than .001) or outpatient psychiatric therapy (3.4% vs. 2.5%, P less than .001).

In proportional hazard models controlling for demographic and clinical factors, receipt of ADT was associated with adjusted hazard ratios of 1.23 for depression and 1.29 (P less than .001 for both) for inpatient psychiatric treatment. There was no significant increase in risk for outpatient psychiatric treatment in this analysis, however.

In addition, the longer patients that were on ADT, the greater the risk for depression. The risk of depression was 12% for patients treated for 6 months or less, 26% for those on ADT for 7-11 months, and 37% for those on ADT for at least 1 year.

“The impact of ADT on depression may plausibly occur via deregulation of neurochemicals, such as serotonin, in addition to the well-described physical effects,” Ms. Dinh and her associates wrote.

Side effects of ADT that can impair quality of life also may contribute to clinical depression, they noted.

The study was supported by charitable grants and internal institutional sources. One investigator reported consulting or advisory roles with Medivation, GenomeDx, and Ferring. Three of the other ten coauthors also reported financial disclosures.

Click for Credit Link
Publications
Topics
Legacy Keywords
ADT
Click for Credit Link
Click for Credit Link

Men on androgen deprivation therapy for prostate cancer are at significantly increased risk for depression, a risk that increases with duration of therapy, investigators report.

A review of Surveillance, Epidemiology, and End Results (SEER) Medicare data on nearly 79,000 men older than 65 years with a diagnosis of prostate cancer showed that those who received androgen deprivation therapy (ADT) had a 23% increased risk for depression, compared with men who were not on ADT, reported Kathryn T. Dinh of Harvard Medical School, Boston, and her colleagues.

“We observed a significantly increased risk of depression and inpatient psychiatric treatment in men treated with ADT for prostate cancer, as well as a duration-response effect such that more ADT was linked to an increasing risk of depression and inpatient and outpatient psychiatric treatment. The possible psychiatric effects of ADT should be recognized by physicians and discussed with patients before initiating treatment,” they wrote (J Clin Oncol. 2016 Apr 11. doi: 10.1200/JCO.2015.64.1969).

 

Although ADT has been identified in some studies as a risk factor for clinical depression, evidence for such a relationship has been spotty, the investigators said, prompting them to conduct a population-based retrospective study to get a better handle on the issue.

They reviewed SEER Medicare data on 78,552 men older than 65 years with a diagnosis of stage I-III prostate cancer treated with ADT from 1992 through 2006, excluding from the sample those patients who had a psychiatric diagnosis within the past 12 months.

Ms. Dinh and her associates found that the 33,882 patients (43%) who received ADT had a significantly higher 3-year cumulative incidence of depression than patients who did not have ADT (7.1% vs. 5.2%, P less than .001), and a significantly higher proportion had either inpatient psychiatric treatment (2.8% vs. 1.9%, P less than .001) or outpatient psychiatric therapy (3.4% vs. 2.5%, P less than .001).

In proportional hazard models controlling for demographic and clinical factors, receipt of ADT was associated with adjusted hazard ratios of 1.23 for depression and 1.29 (P less than .001 for both) for inpatient psychiatric treatment. There was no significant increase in risk for outpatient psychiatric treatment in this analysis, however.

In addition, the longer patients that were on ADT, the greater the risk for depression. The risk of depression was 12% for patients treated for 6 months or less, 26% for those on ADT for 7-11 months, and 37% for those on ADT for at least 1 year.

“The impact of ADT on depression may plausibly occur via deregulation of neurochemicals, such as serotonin, in addition to the well-described physical effects,” Ms. Dinh and her associates wrote.

Side effects of ADT that can impair quality of life also may contribute to clinical depression, they noted.

The study was supported by charitable grants and internal institutional sources. One investigator reported consulting or advisory roles with Medivation, GenomeDx, and Ferring. Three of the other ten coauthors also reported financial disclosures.

Men on androgen deprivation therapy for prostate cancer are at significantly increased risk for depression, a risk that increases with duration of therapy, investigators report.

A review of Surveillance, Epidemiology, and End Results (SEER) Medicare data on nearly 79,000 men older than 65 years with a diagnosis of prostate cancer showed that those who received androgen deprivation therapy (ADT) had a 23% increased risk for depression, compared with men who were not on ADT, reported Kathryn T. Dinh of Harvard Medical School, Boston, and her colleagues.

“We observed a significantly increased risk of depression and inpatient psychiatric treatment in men treated with ADT for prostate cancer, as well as a duration-response effect such that more ADT was linked to an increasing risk of depression and inpatient and outpatient psychiatric treatment. The possible psychiatric effects of ADT should be recognized by physicians and discussed with patients before initiating treatment,” they wrote (J Clin Oncol. 2016 Apr 11. doi: 10.1200/JCO.2015.64.1969).

 

Although ADT has been identified in some studies as a risk factor for clinical depression, evidence for such a relationship has been spotty, the investigators said, prompting them to conduct a population-based retrospective study to get a better handle on the issue.

They reviewed SEER Medicare data on 78,552 men older than 65 years with a diagnosis of stage I-III prostate cancer treated with ADT from 1992 through 2006, excluding from the sample those patients who had a psychiatric diagnosis within the past 12 months.

Ms. Dinh and her associates found that the 33,882 patients (43%) who received ADT had a significantly higher 3-year cumulative incidence of depression than patients who did not have ADT (7.1% vs. 5.2%, P less than .001), and a significantly higher proportion had either inpatient psychiatric treatment (2.8% vs. 1.9%, P less than .001) or outpatient psychiatric therapy (3.4% vs. 2.5%, P less than .001).

In proportional hazard models controlling for demographic and clinical factors, receipt of ADT was associated with adjusted hazard ratios of 1.23 for depression and 1.29 (P less than .001 for both) for inpatient psychiatric treatment. There was no significant increase in risk for outpatient psychiatric treatment in this analysis, however.

In addition, the longer patients that were on ADT, the greater the risk for depression. The risk of depression was 12% for patients treated for 6 months or less, 26% for those on ADT for 7-11 months, and 37% for those on ADT for at least 1 year.

“The impact of ADT on depression may plausibly occur via deregulation of neurochemicals, such as serotonin, in addition to the well-described physical effects,” Ms. Dinh and her associates wrote.

Side effects of ADT that can impair quality of life also may contribute to clinical depression, they noted.

The study was supported by charitable grants and internal institutional sources. One investigator reported consulting or advisory roles with Medivation, GenomeDx, and Ferring. Three of the other ten coauthors also reported financial disclosures.

Publications
Publications
Topics
Article Type
Display Headline
Androgen deprivation therapy linked to depression
Display Headline
Androgen deprivation therapy linked to depression
Legacy Keywords
ADT
Legacy Keywords
ADT
Click for Credit Status
Active
Article Source

FROM THE JOURNAL OF CLINICAL ONCOLOGY

Disallow All Ads
Vitals

Key clinical point: Androgen deprivation therapy for prostate cancer is associated with increased risk for clinical depression.

Major finding: Men who received androgen deprivation therapy (ADT) had a 23% increased risk for depression compared with men who were not on ADT.

Data source: Review of SEER Medicare data on 78,552 men with stage I-III prostate cancer.

Disclosures: The study was supported by charitable grants and internal institutional sources. One investigator reported consulting or advisory roles with Medivation, GenomeDx, and Ferring. Three of the other ten coauthors also reported financial disclosures.

Racial disparities found in HER2-positive breast cancer care

Article Type
Changed
Thu, 12/15/2022 - 17:58
Display Headline
Racial disparities found in HER2-positive breast cancer care

There are significant racial disparities in the delivery of HER2-targeted therapy for breast cancer, investigators have found.

A review of Medicare beneficiaries with incident stage I-III HER2-positive breast cancer showed that black women were 25% less likely than white women to receive adjuvant trastuzumab (Herceptin) within a year of diagnosis, reported Dr. Katherine Reeder-Hayes and her colleagues at the University of North Carolina at Chapel Hill.

In addition, the investigators found that about one-half of Medicare-insured patients with HER2-positive tumors did not receive HER2-targeted therapy, despite the well-known clinical benefits in this population.

“These findings have several troubling implications. First, although this study did not examine survival outcomes, the low overall rates of use among patients with stage II and III disease raise concerns for widespread underuse of trastuzumab in this age group. Second, the racial findings suggest that the presence of a clear biologic predictor of treatment benefit, in this case the HER2 marker, does not mitigate the overall pattern of treatment disparity observed among black women with breast cancer,” they wrote (J Clin Oncol. 2016 April 11. doi: 10.1200/JCO.2015.65.8716).

The authors reviewed Surveillance, Epidemiology and End Results (SEER) Medicare data on 1,362 women with incidence stages I-III HER2-positive breast cancers in 2010 and 2011, and used insurance claims data to identify the use of adjuvant trastuzumab and chemotherapy agents within 12 months of diagnosis. The sample included 1,162 white women, 104 black women, and 96 women described as “other.”

The investigators found that just half of all white women (50%) and 40% of black women received at least some trastuzumab therapy.

In logistic regression analysis controlling for tumor characteristics, socioeconomic factors, and comorbidities, they found that compared with white women, black women had a risk ratio for receiving trastuzumab of 0.745 (P = .0097).

The investigators noted that in addition to the racial disparities, older women and those with more comorbidities were also less likely to get trastuzumab.

“However, given the sizeable benefit of trastuzumab, the overall aggressive biology of this subtype, and extensive literature documenting undertreatment of even healthy older patients, the hypothesis that age is used inappropriately to withhold trastuzumab-based therapy deserves further exploration,” they wrote.

The investigators called for the development of system-level interventions that can objectively identify patients eligible for HER2-targeted therapy, and for ongoing efforts to eradicate racial disparities in care.

The study was supported by grants from the National Institutes of Health and the University (NC) Cancer Research Fund.

References

Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

There are significant racial disparities in the delivery of HER2-targeted therapy for breast cancer, investigators have found.

A review of Medicare beneficiaries with incident stage I-III HER2-positive breast cancer showed that black women were 25% less likely than white women to receive adjuvant trastuzumab (Herceptin) within a year of diagnosis, reported Dr. Katherine Reeder-Hayes and her colleagues at the University of North Carolina at Chapel Hill.

In addition, the investigators found that about one-half of Medicare-insured patients with HER2-positive tumors did not receive HER2-targeted therapy, despite the well-known clinical benefits in this population.

“These findings have several troubling implications. First, although this study did not examine survival outcomes, the low overall rates of use among patients with stage II and III disease raise concerns for widespread underuse of trastuzumab in this age group. Second, the racial findings suggest that the presence of a clear biologic predictor of treatment benefit, in this case the HER2 marker, does not mitigate the overall pattern of treatment disparity observed among black women with breast cancer,” they wrote (J Clin Oncol. 2016 April 11. doi: 10.1200/JCO.2015.65.8716).

The authors reviewed Surveillance, Epidemiology and End Results (SEER) Medicare data on 1,362 women with incidence stages I-III HER2-positive breast cancers in 2010 and 2011, and used insurance claims data to identify the use of adjuvant trastuzumab and chemotherapy agents within 12 months of diagnosis. The sample included 1,162 white women, 104 black women, and 96 women described as “other.”

The investigators found that just half of all white women (50%) and 40% of black women received at least some trastuzumab therapy.

In logistic regression analysis controlling for tumor characteristics, socioeconomic factors, and comorbidities, they found that compared with white women, black women had a risk ratio for receiving trastuzumab of 0.745 (P = .0097).

The investigators noted that in addition to the racial disparities, older women and those with more comorbidities were also less likely to get trastuzumab.

“However, given the sizeable benefit of trastuzumab, the overall aggressive biology of this subtype, and extensive literature documenting undertreatment of even healthy older patients, the hypothesis that age is used inappropriately to withhold trastuzumab-based therapy deserves further exploration,” they wrote.

The investigators called for the development of system-level interventions that can objectively identify patients eligible for HER2-targeted therapy, and for ongoing efforts to eradicate racial disparities in care.

The study was supported by grants from the National Institutes of Health and the University (NC) Cancer Research Fund.

There are significant racial disparities in the delivery of HER2-targeted therapy for breast cancer, investigators have found.

A review of Medicare beneficiaries with incident stage I-III HER2-positive breast cancer showed that black women were 25% less likely than white women to receive adjuvant trastuzumab (Herceptin) within a year of diagnosis, reported Dr. Katherine Reeder-Hayes and her colleagues at the University of North Carolina at Chapel Hill.

In addition, the investigators found that about one-half of Medicare-insured patients with HER2-positive tumors did not receive HER2-targeted therapy, despite the well-known clinical benefits in this population.

“These findings have several troubling implications. First, although this study did not examine survival outcomes, the low overall rates of use among patients with stage II and III disease raise concerns for widespread underuse of trastuzumab in this age group. Second, the racial findings suggest that the presence of a clear biologic predictor of treatment benefit, in this case the HER2 marker, does not mitigate the overall pattern of treatment disparity observed among black women with breast cancer,” they wrote (J Clin Oncol. 2016 April 11. doi: 10.1200/JCO.2015.65.8716).

The authors reviewed Surveillance, Epidemiology and End Results (SEER) Medicare data on 1,362 women with incidence stages I-III HER2-positive breast cancers in 2010 and 2011, and used insurance claims data to identify the use of adjuvant trastuzumab and chemotherapy agents within 12 months of diagnosis. The sample included 1,162 white women, 104 black women, and 96 women described as “other.”

The investigators found that just half of all white women (50%) and 40% of black women received at least some trastuzumab therapy.

In logistic regression analysis controlling for tumor characteristics, socioeconomic factors, and comorbidities, they found that compared with white women, black women had a risk ratio for receiving trastuzumab of 0.745 (P = .0097).

The investigators noted that in addition to the racial disparities, older women and those with more comorbidities were also less likely to get trastuzumab.

“However, given the sizeable benefit of trastuzumab, the overall aggressive biology of this subtype, and extensive literature documenting undertreatment of even healthy older patients, the hypothesis that age is used inappropriately to withhold trastuzumab-based therapy deserves further exploration,” they wrote.

The investigators called for the development of system-level interventions that can objectively identify patients eligible for HER2-targeted therapy, and for ongoing efforts to eradicate racial disparities in care.

The study was supported by grants from the National Institutes of Health and the University (NC) Cancer Research Fund.

References

References

Publications
Publications
Topics
Article Type
Display Headline
Racial disparities found in HER2-positive breast cancer care
Display Headline
Racial disparities found in HER2-positive breast cancer care
Article Source

FROM THE JOURNAL OF CLINICAL ONCOLOGY

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Trastuzumab is known to be beneficial in women with HER2-positive breast cancer, but black women were less likely to receive it than white women.

Major finding: Only 50% of white women and 40% of black women with HER2-positive cancers received trastuzumab.

Data source: A retrospective study of SEER Medicare data on 1,362 women with stage I-III HER2-positive breast cancer.

Disclosures: The study was supported by grants from the National Institutes of Health and the University (NC) Cancer Research Fund.

Guidelines highlight revolution in advanced melanoma therapies

Article Type
Changed
Mon, 01/14/2019 - 09:36
Display Headline
Guidelines highlight revolution in advanced melanoma therapies

HOLLYWOOD, FLA. –The remarkable changes that have occurred in the treatment of advanced malignant melanoma over the last several years are reflected in revised National Comprehensive Cancer Network guidelines.

“Gone are the days where our active agents were interferon and dacarbazine,” said Dr. John A. Thompson of the Fred Hutchinson Cancer Research Center, Seattle.

Dr. John A. Thompson

“We’ve seen the approval by the FDA of the PD-1-targeted agents and also molecular-targeted agents to treat patients with mutations in the BRAF gene, and the rate of change continues apace,” he said at the annual conference of the National Comprehensive Cancer Network.

Among the changes in the 2016 guidelines are the addition of high-dose ipilimumab (Yervoy) as an option for stage III (node-positive) disease, and intralesional talimogene laherparepvec (T-VEC) for primary treatment of stage III in-transit disease.

Guidelines for first-line systemic therapy for metastatic or unresectable disease recommend treatment based on evaluation of individual patients and include:

• Monotherapy with an anti-PD-1 (programmed death-1) agent, either pembrolizumab (Keytruda) or nivolumab (Opdivo) (category 1 recommendation).

• Targeted therapy for BRAF-mutated disease with combinations (preferred) of either dabrafenib (Tafinlar) and trametinib (Mekinist) or vemurafenib (Zelboraf)and cobimetinib (Cotellic); both are category 1 recommendations.

• Single-agent therapy with either vemurafenib or dabrafenib (category 1), or clinical trial.

For patients with disease progression or those who have had their maximum clinical benefit from BRAF-targeted therapy and good performance status (0-2), the guidelines recommend second-line or subsequent therapy with anti-PD-1 agents as above, ipilimumab, or a combination of nivolumab and ipilimumab.

The guidelines note, however, that “nivolumab/ipilimumab combination therapy is associated with improved relapse-free survival compared with single-agent nivolumab or ipilimumab, at the expense of significantly increased toxicity. Compared to single-agent therapy, the impact of nivolumab/ipilimumab combination therapy on overall survival is not known.”

Also recommended are targeted therapy with the BRAF-inhibitor combinations listed above (preferred) or single-agent therapy with vemurafenib or dabrafenib.

“In previously untreated patients with unresectable stage IIIC or stage IV disease, the combination of vemurafenib/cobimetinib was associated with improved PFS [progression-free survival] and response rate when compared to vemurafenib alone. The impact on overall survival compared to single-agent vemurafenib is unknown,” the guidelines state.

Other second-line options for patients with good performance status include high-dose interleukin-2 (not suitable for patients with inadequate organ reserve, poor performance status, or untreated or active brain metastases), biochemotherapy and cytotoxic agents (dacarbazine or temozolomide, and cisplatin or carboplatin, with or without vinblastine or nitrosourea, and IL-2 and interferon alfa-2b; paclitaxel, albumin-bound paclitaxel).

For patients with poor performance status (3-4) the guidelines call for best supportive care according to guidelines for palliative care.

Oncolytic immunotherapy

For treatment of recurrent disease, the guidelines newly recommend use of T-VEC, a genetically modified herpes simplex-1 virus that is injected intralesionally into accessible tumors. The attenuated virus does not kill healthy cells, but replicates in tumors and causes the cells to secrete granulocyte macrophage–colony stimulating factor (GM-CSF), leading to lysis of the tumor cells.

“It is hypothesized that the GM-CSF works with the immune system to boost a resident immune response against the destroyed melanoma cells and antigens that are released from this process,” Dr. Thompson said.

In a recent clinical trial (J Clin Oncol. 2015;33:2780-8), patients with injectable melanoma that was not surgically resectable were randomized to either intralesional T-VEC injections on week 0, 3, and then every 2 weeks (295 patients), or subcutaneous GM-CSF 125 mcg/m2 on days 1 to 14 for every 28 days (141 patients).

T-VEC was associated with significantly more durables responses (16% vs. 2.1% for GM-CSF, P less than .001) and a significantly higher overall response rate (26.4% vs. 5.7%, respectively, P less than .001).

In the primary analysis in an intent-to-treat population, T-VEC had a borderline significant association with better overall survival (median 23.2 vs. 18.9 months, hazard ratio [HR] 0.70, log-rank P = .051).

In subanalyses of outcomes in patient subgroups, however, the benefits of T-VEC were seen in patients with stage IIIB IIIC, and IV M1a disease (HR 0.57, P less than .001), but not in patients with IV M1b or M1c disease. In addition, patients treated with T-VEC in first-line therapy had significantly better overall survival compared with GM-CSF (HR 0.50, P less than .001), but those who received it in second or subsequent lines of therapy saw no survival benefit from intralesional injection.

Future directions for the treatment of advanced melanoma include identification of predictive biomarkers to guide the choice of therapy, new immune agonist antibodies or checkpoint inhibitors to act in concert with PD-1 inhibitors, adaptive T-cell therapies, and lymphokines, cytokines, vaccines, and targeted therapies that can overcome mechanisms of resistance and work in combination with immunomodulators, Dr. Thompson said.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

HOLLYWOOD, FLA. –The remarkable changes that have occurred in the treatment of advanced malignant melanoma over the last several years are reflected in revised National Comprehensive Cancer Network guidelines.

“Gone are the days where our active agents were interferon and dacarbazine,” said Dr. John A. Thompson of the Fred Hutchinson Cancer Research Center, Seattle.

Dr. John A. Thompson

“We’ve seen the approval by the FDA of the PD-1-targeted agents and also molecular-targeted agents to treat patients with mutations in the BRAF gene, and the rate of change continues apace,” he said at the annual conference of the National Comprehensive Cancer Network.

Among the changes in the 2016 guidelines are the addition of high-dose ipilimumab (Yervoy) as an option for stage III (node-positive) disease, and intralesional talimogene laherparepvec (T-VEC) for primary treatment of stage III in-transit disease.

Guidelines for first-line systemic therapy for metastatic or unresectable disease recommend treatment based on evaluation of individual patients and include:

• Monotherapy with an anti-PD-1 (programmed death-1) agent, either pembrolizumab (Keytruda) or nivolumab (Opdivo) (category 1 recommendation).

• Targeted therapy for BRAF-mutated disease with combinations (preferred) of either dabrafenib (Tafinlar) and trametinib (Mekinist) or vemurafenib (Zelboraf)and cobimetinib (Cotellic); both are category 1 recommendations.

• Single-agent therapy with either vemurafenib or dabrafenib (category 1), or clinical trial.

For patients with disease progression or those who have had their maximum clinical benefit from BRAF-targeted therapy and good performance status (0-2), the guidelines recommend second-line or subsequent therapy with anti-PD-1 agents as above, ipilimumab, or a combination of nivolumab and ipilimumab.

The guidelines note, however, that “nivolumab/ipilimumab combination therapy is associated with improved relapse-free survival compared with single-agent nivolumab or ipilimumab, at the expense of significantly increased toxicity. Compared to single-agent therapy, the impact of nivolumab/ipilimumab combination therapy on overall survival is not known.”

Also recommended are targeted therapy with the BRAF-inhibitor combinations listed above (preferred) or single-agent therapy with vemurafenib or dabrafenib.

“In previously untreated patients with unresectable stage IIIC or stage IV disease, the combination of vemurafenib/cobimetinib was associated with improved PFS [progression-free survival] and response rate when compared to vemurafenib alone. The impact on overall survival compared to single-agent vemurafenib is unknown,” the guidelines state.

Other second-line options for patients with good performance status include high-dose interleukin-2 (not suitable for patients with inadequate organ reserve, poor performance status, or untreated or active brain metastases), biochemotherapy and cytotoxic agents (dacarbazine or temozolomide, and cisplatin or carboplatin, with or without vinblastine or nitrosourea, and IL-2 and interferon alfa-2b; paclitaxel, albumin-bound paclitaxel).

For patients with poor performance status (3-4) the guidelines call for best supportive care according to guidelines for palliative care.

Oncolytic immunotherapy

For treatment of recurrent disease, the guidelines newly recommend use of T-VEC, a genetically modified herpes simplex-1 virus that is injected intralesionally into accessible tumors. The attenuated virus does not kill healthy cells, but replicates in tumors and causes the cells to secrete granulocyte macrophage–colony stimulating factor (GM-CSF), leading to lysis of the tumor cells.

“It is hypothesized that the GM-CSF works with the immune system to boost a resident immune response against the destroyed melanoma cells and antigens that are released from this process,” Dr. Thompson said.

In a recent clinical trial (J Clin Oncol. 2015;33:2780-8), patients with injectable melanoma that was not surgically resectable were randomized to either intralesional T-VEC injections on week 0, 3, and then every 2 weeks (295 patients), or subcutaneous GM-CSF 125 mcg/m2 on days 1 to 14 for every 28 days (141 patients).

T-VEC was associated with significantly more durables responses (16% vs. 2.1% for GM-CSF, P less than .001) and a significantly higher overall response rate (26.4% vs. 5.7%, respectively, P less than .001).

In the primary analysis in an intent-to-treat population, T-VEC had a borderline significant association with better overall survival (median 23.2 vs. 18.9 months, hazard ratio [HR] 0.70, log-rank P = .051).

In subanalyses of outcomes in patient subgroups, however, the benefits of T-VEC were seen in patients with stage IIIB IIIC, and IV M1a disease (HR 0.57, P less than .001), but not in patients with IV M1b or M1c disease. In addition, patients treated with T-VEC in first-line therapy had significantly better overall survival compared with GM-CSF (HR 0.50, P less than .001), but those who received it in second or subsequent lines of therapy saw no survival benefit from intralesional injection.

Future directions for the treatment of advanced melanoma include identification of predictive biomarkers to guide the choice of therapy, new immune agonist antibodies or checkpoint inhibitors to act in concert with PD-1 inhibitors, adaptive T-cell therapies, and lymphokines, cytokines, vaccines, and targeted therapies that can overcome mechanisms of resistance and work in combination with immunomodulators, Dr. Thompson said.

HOLLYWOOD, FLA. –The remarkable changes that have occurred in the treatment of advanced malignant melanoma over the last several years are reflected in revised National Comprehensive Cancer Network guidelines.

“Gone are the days where our active agents were interferon and dacarbazine,” said Dr. John A. Thompson of the Fred Hutchinson Cancer Research Center, Seattle.

Dr. John A. Thompson

“We’ve seen the approval by the FDA of the PD-1-targeted agents and also molecular-targeted agents to treat patients with mutations in the BRAF gene, and the rate of change continues apace,” he said at the annual conference of the National Comprehensive Cancer Network.

Among the changes in the 2016 guidelines are the addition of high-dose ipilimumab (Yervoy) as an option for stage III (node-positive) disease, and intralesional talimogene laherparepvec (T-VEC) for primary treatment of stage III in-transit disease.

Guidelines for first-line systemic therapy for metastatic or unresectable disease recommend treatment based on evaluation of individual patients and include:

• Monotherapy with an anti-PD-1 (programmed death-1) agent, either pembrolizumab (Keytruda) or nivolumab (Opdivo) (category 1 recommendation).

• Targeted therapy for BRAF-mutated disease with combinations (preferred) of either dabrafenib (Tafinlar) and trametinib (Mekinist) or vemurafenib (Zelboraf)and cobimetinib (Cotellic); both are category 1 recommendations.

• Single-agent therapy with either vemurafenib or dabrafenib (category 1), or clinical trial.

For patients with disease progression or those who have had their maximum clinical benefit from BRAF-targeted therapy and good performance status (0-2), the guidelines recommend second-line or subsequent therapy with anti-PD-1 agents as above, ipilimumab, or a combination of nivolumab and ipilimumab.

The guidelines note, however, that “nivolumab/ipilimumab combination therapy is associated with improved relapse-free survival compared with single-agent nivolumab or ipilimumab, at the expense of significantly increased toxicity. Compared to single-agent therapy, the impact of nivolumab/ipilimumab combination therapy on overall survival is not known.”

Also recommended are targeted therapy with the BRAF-inhibitor combinations listed above (preferred) or single-agent therapy with vemurafenib or dabrafenib.

“In previously untreated patients with unresectable stage IIIC or stage IV disease, the combination of vemurafenib/cobimetinib was associated with improved PFS [progression-free survival] and response rate when compared to vemurafenib alone. The impact on overall survival compared to single-agent vemurafenib is unknown,” the guidelines state.

Other second-line options for patients with good performance status include high-dose interleukin-2 (not suitable for patients with inadequate organ reserve, poor performance status, or untreated or active brain metastases), biochemotherapy and cytotoxic agents (dacarbazine or temozolomide, and cisplatin or carboplatin, with or without vinblastine or nitrosourea, and IL-2 and interferon alfa-2b; paclitaxel, albumin-bound paclitaxel).

For patients with poor performance status (3-4) the guidelines call for best supportive care according to guidelines for palliative care.

Oncolytic immunotherapy

For treatment of recurrent disease, the guidelines newly recommend use of T-VEC, a genetically modified herpes simplex-1 virus that is injected intralesionally into accessible tumors. The attenuated virus does not kill healthy cells, but replicates in tumors and causes the cells to secrete granulocyte macrophage–colony stimulating factor (GM-CSF), leading to lysis of the tumor cells.

“It is hypothesized that the GM-CSF works with the immune system to boost a resident immune response against the destroyed melanoma cells and antigens that are released from this process,” Dr. Thompson said.

In a recent clinical trial (J Clin Oncol. 2015;33:2780-8), patients with injectable melanoma that was not surgically resectable were randomized to either intralesional T-VEC injections on week 0, 3, and then every 2 weeks (295 patients), or subcutaneous GM-CSF 125 mcg/m2 on days 1 to 14 for every 28 days (141 patients).

T-VEC was associated with significantly more durables responses (16% vs. 2.1% for GM-CSF, P less than .001) and a significantly higher overall response rate (26.4% vs. 5.7%, respectively, P less than .001).

In the primary analysis in an intent-to-treat population, T-VEC had a borderline significant association with better overall survival (median 23.2 vs. 18.9 months, hazard ratio [HR] 0.70, log-rank P = .051).

In subanalyses of outcomes in patient subgroups, however, the benefits of T-VEC were seen in patients with stage IIIB IIIC, and IV M1a disease (HR 0.57, P less than .001), but not in patients with IV M1b or M1c disease. In addition, patients treated with T-VEC in first-line therapy had significantly better overall survival compared with GM-CSF (HR 0.50, P less than .001), but those who received it in second or subsequent lines of therapy saw no survival benefit from intralesional injection.

Future directions for the treatment of advanced melanoma include identification of predictive biomarkers to guide the choice of therapy, new immune agonist antibodies or checkpoint inhibitors to act in concert with PD-1 inhibitors, adaptive T-cell therapies, and lymphokines, cytokines, vaccines, and targeted therapies that can overcome mechanisms of resistance and work in combination with immunomodulators, Dr. Thompson said.

References

References

Publications
Publications
Topics
Article Type
Display Headline
Guidelines highlight revolution in advanced melanoma therapies
Display Headline
Guidelines highlight revolution in advanced melanoma therapies
Article Source

AT THE NCCN ANNUAL CONFERENCE

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Immunotherapies and targeted agents have revolutionized the treatment of advanced melanoma.

Major finding: Current therapies are associated with response rates ranging from 60% to 75%.

Data source: Review of NCCN melanoma guideline changes and evidence supporting the changes.

Disclosures: Dr. Thompson disclosed consulting fees or honoraria from Eisai, Genentech, and Seattle Genetics, and “other financial benefit” from Celidex Therapeutics.

New vulvar cancer guidelines stress regional disease control

Article Type
Changed
Fri, 01/04/2019 - 13:14
Display Headline
New vulvar cancer guidelines stress regional disease control

HOLLYWOOD, FLA. – The National Comprehensive Cancer Network has issued new guidelines for the diagnosis and management of vulvar cancer.

Vulvar cancers are rare neoplasms, with an estimated U.S. annual incidence of 5,950 cases, and 1,110 deaths. The majority of cases (about 90%) are of squamous cell histology.

Dr. Benjamin E. Greer

Treatment of vulvar cancer has evolved from en bloc resections used throughout most of the 20th century, to more refined techniques, said Dr. Benjamin E. Greer, professor of gynecological oncology at the University of Washington in Seattle.

“In the 1980s, we started to modify treatment to reduce morbidity,” he said at the annual conference of the National Comprehensive Cancer Network.

With older, more radical techniques, groin breakdown, leg edema, and impaired sexual function were common post-surgery consequences. Current practice, however, is to perform regional lymph node management for unilateral cancers, radical local excision rather than en bloc resections, separate groin incisions, lymphatic mapping, radiation, chemotherapy, and, if necessary, exenteration, Dr. Greer noted.

The guidelines note that adequate surgical margins – 1 to 2 cm – at the time of primary surgery appear to be essential for reducing risk of local recurrence, and that if margins are within 8 mm of tumor, the surgeon should consider re-excision or adjuvant radiation.

Lymph node status is the most important determinant of survival, with historical reports showing overall survival following surgery of 70% to 80% among patients with negative nodes, compared with 30% to 40% of those with positive nodes, he said.

Evaluation of bilateral inguinofemoral groin nodes should be performed in patients with lesions in the vulvar midline, and ipsilateral groin node evaluation should be performed for those with lateral lesions lying more than 2 cm from the vulvar midline. Additionally, select patients may require sentinel lymph node biopsy, the guidelines state.

Unilateral carcinomas of the vulva can be treated with limited radical vulvectomy and ipsilateral inguinal femoral node dissection. Lymph node dissection can be performed through a separate incision. For patients with positive nodes, adjuvant radiation may aid in disease control. Patients with inoperable carcinomas are recommended to receive radiation and chemotherapy.

Radiation for vulvar cancer

“For early stage tumors, adjuvant radiotherapy is an effective treatment modality that significantly decreases recurrence, especially in surgically resected groins, and it leads to improvement in relapse-free and overall survival,” said Dr. Wui-Jin Koh, medical director for radiation oncology at the Fred Hutchinson Cancer Research Center in Seattle.

Concurrent chemotherapy and radiation may provide additional therapeutic benefit, especially for patients with advanced, unresectable tumors, and it may help to address systemic risk in patients with multiple positive lymph nodes, Dr. Koh said.

Dr. Wui-Jin Koh

The guidelines state that radiation can be given with external beam radiation delivered via a 3D-conformal or intensity modulated (IMRT) technique, with brachytherapy boost for some tumors where the anatomy permits.

“Careful attention should be taken to ensure adequate tumor coverage by combining clinical examination, imaging findings, and appropriate nodal volumes at risk to define the target volume,” the guideline states.

For adjuvant therapy, doses of 50.4 Gy divided in 1.8 Gy fractions should be delivered once daily 5 days per week, with minimal treatment breaks.

For treatment of unresectable tumors, doses range from 59.4 Gy to 64.8 Gy in 1.8 Gy fractions, with a boost dose to approximately 70 Gy for large lymph nodes in select cases.

Residual disease

The decision to provide additional treatment following surgery is based on whether the patient is clinically negative for residual tumor at the primary site and nodes.

“If one has negative margins and negative nodes? Observation, absolutely,” Dr. Koh said. “If one has positive margins for invasive disease, our recommendation is to re-excise and not go straight to radiation, and if one can do it and get negative margins, again observe the majority of them.”

“Use radiation very judiciously,” he added. “Only if patients have positive margins or have unresectable primary disease do we routinely recommend radiation.”

Locally advanced disease

For patients who cannot be treated with conventional or sphincter-sparing, organ preserving surgery upfront, the recommendation is to provide chemoradiation, with initial radiation to the primary site, groins, and pelvis, and concurrent week cisplatin at a dose of 30-40 mg/m2 per week. The recommended radiation doses are 45 Gy to at-risk, microscopic clinical tumor volume, and 57.6 to 60 Gy to gross tumor volume (primary site and nodes).

“If one uses IMRT, you need to be very generous with the volumes,” Dr. Koh said.

The panelists also recommend re-imaging and re-evaluating patients 6 to 8 weeks after the completion of chemoradiation, with possible resection or biopsy of the primary tumor site, and limited groin resection of imaged residual disease.

 

 

For patients with clearly node-positive disease, “my general preference is to give upfront chemoradiation therapy to avoid delay of primary therapy, and then resect residual nodes after the chemoradiation is done,” he said.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

HOLLYWOOD, FLA. – The National Comprehensive Cancer Network has issued new guidelines for the diagnosis and management of vulvar cancer.

Vulvar cancers are rare neoplasms, with an estimated U.S. annual incidence of 5,950 cases, and 1,110 deaths. The majority of cases (about 90%) are of squamous cell histology.

Dr. Benjamin E. Greer

Treatment of vulvar cancer has evolved from en bloc resections used throughout most of the 20th century, to more refined techniques, said Dr. Benjamin E. Greer, professor of gynecological oncology at the University of Washington in Seattle.

“In the 1980s, we started to modify treatment to reduce morbidity,” he said at the annual conference of the National Comprehensive Cancer Network.

With older, more radical techniques, groin breakdown, leg edema, and impaired sexual function were common post-surgery consequences. Current practice, however, is to perform regional lymph node management for unilateral cancers, radical local excision rather than en bloc resections, separate groin incisions, lymphatic mapping, radiation, chemotherapy, and, if necessary, exenteration, Dr. Greer noted.

The guidelines note that adequate surgical margins – 1 to 2 cm – at the time of primary surgery appear to be essential for reducing risk of local recurrence, and that if margins are within 8 mm of tumor, the surgeon should consider re-excision or adjuvant radiation.

Lymph node status is the most important determinant of survival, with historical reports showing overall survival following surgery of 70% to 80% among patients with negative nodes, compared with 30% to 40% of those with positive nodes, he said.

Evaluation of bilateral inguinofemoral groin nodes should be performed in patients with lesions in the vulvar midline, and ipsilateral groin node evaluation should be performed for those with lateral lesions lying more than 2 cm from the vulvar midline. Additionally, select patients may require sentinel lymph node biopsy, the guidelines state.

Unilateral carcinomas of the vulva can be treated with limited radical vulvectomy and ipsilateral inguinal femoral node dissection. Lymph node dissection can be performed through a separate incision. For patients with positive nodes, adjuvant radiation may aid in disease control. Patients with inoperable carcinomas are recommended to receive radiation and chemotherapy.

Radiation for vulvar cancer

“For early stage tumors, adjuvant radiotherapy is an effective treatment modality that significantly decreases recurrence, especially in surgically resected groins, and it leads to improvement in relapse-free and overall survival,” said Dr. Wui-Jin Koh, medical director for radiation oncology at the Fred Hutchinson Cancer Research Center in Seattle.

Concurrent chemotherapy and radiation may provide additional therapeutic benefit, especially for patients with advanced, unresectable tumors, and it may help to address systemic risk in patients with multiple positive lymph nodes, Dr. Koh said.

Dr. Wui-Jin Koh

The guidelines state that radiation can be given with external beam radiation delivered via a 3D-conformal or intensity modulated (IMRT) technique, with brachytherapy boost for some tumors where the anatomy permits.

“Careful attention should be taken to ensure adequate tumor coverage by combining clinical examination, imaging findings, and appropriate nodal volumes at risk to define the target volume,” the guideline states.

For adjuvant therapy, doses of 50.4 Gy divided in 1.8 Gy fractions should be delivered once daily 5 days per week, with minimal treatment breaks.

For treatment of unresectable tumors, doses range from 59.4 Gy to 64.8 Gy in 1.8 Gy fractions, with a boost dose to approximately 70 Gy for large lymph nodes in select cases.

Residual disease

The decision to provide additional treatment following surgery is based on whether the patient is clinically negative for residual tumor at the primary site and nodes.

“If one has negative margins and negative nodes? Observation, absolutely,” Dr. Koh said. “If one has positive margins for invasive disease, our recommendation is to re-excise and not go straight to radiation, and if one can do it and get negative margins, again observe the majority of them.”

“Use radiation very judiciously,” he added. “Only if patients have positive margins or have unresectable primary disease do we routinely recommend radiation.”

Locally advanced disease

For patients who cannot be treated with conventional or sphincter-sparing, organ preserving surgery upfront, the recommendation is to provide chemoradiation, with initial radiation to the primary site, groins, and pelvis, and concurrent week cisplatin at a dose of 30-40 mg/m2 per week. The recommended radiation doses are 45 Gy to at-risk, microscopic clinical tumor volume, and 57.6 to 60 Gy to gross tumor volume (primary site and nodes).

“If one uses IMRT, you need to be very generous with the volumes,” Dr. Koh said.

The panelists also recommend re-imaging and re-evaluating patients 6 to 8 weeks after the completion of chemoradiation, with possible resection or biopsy of the primary tumor site, and limited groin resection of imaged residual disease.

 

 

For patients with clearly node-positive disease, “my general preference is to give upfront chemoradiation therapy to avoid delay of primary therapy, and then resect residual nodes after the chemoradiation is done,” he said.

HOLLYWOOD, FLA. – The National Comprehensive Cancer Network has issued new guidelines for the diagnosis and management of vulvar cancer.

Vulvar cancers are rare neoplasms, with an estimated U.S. annual incidence of 5,950 cases, and 1,110 deaths. The majority of cases (about 90%) are of squamous cell histology.

Dr. Benjamin E. Greer

Treatment of vulvar cancer has evolved from en bloc resections used throughout most of the 20th century, to more refined techniques, said Dr. Benjamin E. Greer, professor of gynecological oncology at the University of Washington in Seattle.

“In the 1980s, we started to modify treatment to reduce morbidity,” he said at the annual conference of the National Comprehensive Cancer Network.

With older, more radical techniques, groin breakdown, leg edema, and impaired sexual function were common post-surgery consequences. Current practice, however, is to perform regional lymph node management for unilateral cancers, radical local excision rather than en bloc resections, separate groin incisions, lymphatic mapping, radiation, chemotherapy, and, if necessary, exenteration, Dr. Greer noted.

The guidelines note that adequate surgical margins – 1 to 2 cm – at the time of primary surgery appear to be essential for reducing risk of local recurrence, and that if margins are within 8 mm of tumor, the surgeon should consider re-excision or adjuvant radiation.

Lymph node status is the most important determinant of survival, with historical reports showing overall survival following surgery of 70% to 80% among patients with negative nodes, compared with 30% to 40% of those with positive nodes, he said.

Evaluation of bilateral inguinofemoral groin nodes should be performed in patients with lesions in the vulvar midline, and ipsilateral groin node evaluation should be performed for those with lateral lesions lying more than 2 cm from the vulvar midline. Additionally, select patients may require sentinel lymph node biopsy, the guidelines state.

Unilateral carcinomas of the vulva can be treated with limited radical vulvectomy and ipsilateral inguinal femoral node dissection. Lymph node dissection can be performed through a separate incision. For patients with positive nodes, adjuvant radiation may aid in disease control. Patients with inoperable carcinomas are recommended to receive radiation and chemotherapy.

Radiation for vulvar cancer

“For early stage tumors, adjuvant radiotherapy is an effective treatment modality that significantly decreases recurrence, especially in surgically resected groins, and it leads to improvement in relapse-free and overall survival,” said Dr. Wui-Jin Koh, medical director for radiation oncology at the Fred Hutchinson Cancer Research Center in Seattle.

Concurrent chemotherapy and radiation may provide additional therapeutic benefit, especially for patients with advanced, unresectable tumors, and it may help to address systemic risk in patients with multiple positive lymph nodes, Dr. Koh said.

Dr. Wui-Jin Koh

The guidelines state that radiation can be given with external beam radiation delivered via a 3D-conformal or intensity modulated (IMRT) technique, with brachytherapy boost for some tumors where the anatomy permits.

“Careful attention should be taken to ensure adequate tumor coverage by combining clinical examination, imaging findings, and appropriate nodal volumes at risk to define the target volume,” the guideline states.

For adjuvant therapy, doses of 50.4 Gy divided in 1.8 Gy fractions should be delivered once daily 5 days per week, with minimal treatment breaks.

For treatment of unresectable tumors, doses range from 59.4 Gy to 64.8 Gy in 1.8 Gy fractions, with a boost dose to approximately 70 Gy for large lymph nodes in select cases.

Residual disease

The decision to provide additional treatment following surgery is based on whether the patient is clinically negative for residual tumor at the primary site and nodes.

“If one has negative margins and negative nodes? Observation, absolutely,” Dr. Koh said. “If one has positive margins for invasive disease, our recommendation is to re-excise and not go straight to radiation, and if one can do it and get negative margins, again observe the majority of them.”

“Use radiation very judiciously,” he added. “Only if patients have positive margins or have unresectable primary disease do we routinely recommend radiation.”

Locally advanced disease

For patients who cannot be treated with conventional or sphincter-sparing, organ preserving surgery upfront, the recommendation is to provide chemoradiation, with initial radiation to the primary site, groins, and pelvis, and concurrent week cisplatin at a dose of 30-40 mg/m2 per week. The recommended radiation doses are 45 Gy to at-risk, microscopic clinical tumor volume, and 57.6 to 60 Gy to gross tumor volume (primary site and nodes).

“If one uses IMRT, you need to be very generous with the volumes,” Dr. Koh said.

The panelists also recommend re-imaging and re-evaluating patients 6 to 8 weeks after the completion of chemoradiation, with possible resection or biopsy of the primary tumor site, and limited groin resection of imaged residual disease.

 

 

For patients with clearly node-positive disease, “my general preference is to give upfront chemoradiation therapy to avoid delay of primary therapy, and then resect residual nodes after the chemoradiation is done,” he said.

References

References

Publications
Publications
Topics
Article Type
Display Headline
New vulvar cancer guidelines stress regional disease control
Display Headline
New vulvar cancer guidelines stress regional disease control
Sections
Article Source

AT THE NCCN ANNUAL CONFERENCE

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Nodal status is an important determinant of survival of patients with vulvar carcinomas.

Major finding: Historically, reported overall survival following surgery is 70% to 80% among patients with negative nodes, compared with 30% to 40% of those with positive nodes.

Data source: Review of new clinical guidelines for the management of patients with vulvar cancer.

Disclosures: Dr. Greer and Dr. Koh reported having no relevant clinical disclosures.

NCCN breast cancer RT guidelines: hypofractionation preferred

Article Type
Changed
Thu, 12/15/2022 - 17:58
Display Headline
NCCN breast cancer RT guidelines: hypofractionation preferred

HOLLYWOOD, FLA. – No huge surprises here, but newly revised National Comprehensive Cancer Network guidelines for locoregional therapy of breast cancer help clarify the optimal use of adjuvant radiation.

For example, updates to the pages on principles of radiation therapy in invasive breast cancer contain new information on optimal dosing, fractionation, treatment planning, and techniques for minimizing radiation of surrounding normal tissues, noted Dr. Kilian E. Salerno, director of breast radiation and soft tissue/melanoma radiation at the Roswell Park Cancer Institute in Buffalo, New York.

Dr. Kilian E. Salerno

“There are a number of treatment options, many more now than ever before,” she said at the annual conference of the National Comprehensive Cancer Network.

The guidelines emphasize the importance of individualized radiation therapy planning and delivery. A CT-based treatment plan can help clinicians choose target volumes and identify adjacent organs and tissues that may be at risk for radiation spill.

Recommended options for reducing radiation doses to adjacent tissues include deep breath holds and prone positioning so that the treated breast hangs down, helping to isolate it from the heart and lungs.

Whole breast irradiation

Whole breast irradiation is the standard for radiation therapy of early breast cancer following breast-conserving surgery, although partial breast irradiation, typically of lumpectomy cavities, is also commonly performed, Dr. Salerno said.

Boost radiation in conjunction with breast conserving therapy can be delivered via en face electron fields, photons, or brachytherapy; boost dosing to chest wall scar is usually treated with photons or electrons, the guidelines note.

The guidelines recommend weekly imaging to ensure that daily treatment setup is consistent, but caution against routine use of daily imaging.

For whole breast irradiation, the guidelines recommend either doses of 46-50 Gy divided into 23-25 fractions (conventional fractionation), or, preferably, hypofractionated radiation delivered in doses of 40-42.5 Gy over 15-16 fractions, with all doses given 5 days per week. Patients at higher risk for recurrence should receive boost doses to the tumor bed, typically at doses of 10-16 Gy delivered in 4-8 fractions.

The guidelines now favor hypofractionation based on long-term results from clinical trials conducted in Ontario, Canada, and in London.

In the Canadian trial (N Engl J Med. 2010;362:513-20), patients were treated with whole-breast irradiation at doses of 42.5 Gy in 16 fractions, with no boost dose.

In the Standardisation of Breast Radiotherapy B (START B) trial (Lancet Oncol. 14;11:1086-94), in London, patients received 40 Gy in 15 fractions without a boost dose.

Both trials showed that hypofractionation was associated with disease outcomes that were equivalent or better to those seen with conventional fractionation schedules, as well as equivalent or better cosmesis and adverse event profiles, Dr. Salerno said.

“This is for whole breast radiation; it is not to be routinely used in the post-mastectomy setting or when you’re treating regional nodes,” she said.

Accelerated partial breast irradiation

For accelerated partial breast irradiation (APBI), the guidelines recommend a hypofractionated schedule of 34 Gy divided into 10 fractions delivered twice daily for 5 days for brachytherapy, or 38.5 Gy in 10 twice daily fractions with external beam therapy to the tumor bed.

The NCCN guidelines for APBI are based on the American Society of Radiation Oncology (ASTRO) consensus statement for APBI. An updated statement is planned for 2017.

Omit radiation?

Results from the CALGB 9343 trial and other studies suggest that in selected women with lower risk for recurrence, radiation can be omitted without compromising survival. These include women aged 70 or older with small primary breast cancers, negative nodes, negative surgical margins, and hormone-receptor-positive disease who are treated with breast conserving surgery and go on to tamoxifen maintenance.

Post-mastectomy radiation

Following total mastectomy with surgical axillary staging, with our without reconstruction, the guidelines recommend specific steps based on the number of nodes involved and the tumor size and/or margin status.

Thus, women with four or more positive axillary nodes should receive radiation to the chest wall and infraclavicular region, supraclavicular area, internal mammary nodes, and any at-risk part of the axillary bed. If adjuvant chemotherapy is prescribed, radiation usually is given following chemotherapy completion.

For women with one to three positive axillary nodes, the guidelines urge clinicians to “strongly consider” treating them as they would women with four or more positive nodes. The same advice applies to women with negative nodes but tumors larger than 5 cm or positive surgical margins as defined by ink on tumor.

For patients with negative nodes, tumors 5 cm or smaller and negative but narrow margins (less than 1 mm), the guidelines recommend consideration of radiation to the chest wall. Patients with small tumors, no nodal involvement, and margins 1 mm or greater can be spared radiation therapy.

 

 

Recurrence

For women with local-only recurrence, the guidelines recommend mastectomy with axillary lymph node staging for those with recurrences after initial lumpectomy and radiation, or if the initial treatment was mastectomy with no radiation, surgical resection with radiation, or surgical resection alone for those women previously treated with mastectomy and radiation.

For regional or locoregional recurrences, the guidelines recommend that axillary recurrences be treated with surgery and radiation if possible, and that supraclavicular or internal mammary node recurrences be treated with radiation therapy, if possible.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

HOLLYWOOD, FLA. – No huge surprises here, but newly revised National Comprehensive Cancer Network guidelines for locoregional therapy of breast cancer help clarify the optimal use of adjuvant radiation.

For example, updates to the pages on principles of radiation therapy in invasive breast cancer contain new information on optimal dosing, fractionation, treatment planning, and techniques for minimizing radiation of surrounding normal tissues, noted Dr. Kilian E. Salerno, director of breast radiation and soft tissue/melanoma radiation at the Roswell Park Cancer Institute in Buffalo, New York.

Dr. Kilian E. Salerno

“There are a number of treatment options, many more now than ever before,” she said at the annual conference of the National Comprehensive Cancer Network.

The guidelines emphasize the importance of individualized radiation therapy planning and delivery. A CT-based treatment plan can help clinicians choose target volumes and identify adjacent organs and tissues that may be at risk for radiation spill.

Recommended options for reducing radiation doses to adjacent tissues include deep breath holds and prone positioning so that the treated breast hangs down, helping to isolate it from the heart and lungs.

Whole breast irradiation

Whole breast irradiation is the standard for radiation therapy of early breast cancer following breast-conserving surgery, although partial breast irradiation, typically of lumpectomy cavities, is also commonly performed, Dr. Salerno said.

Boost radiation in conjunction with breast conserving therapy can be delivered via en face electron fields, photons, or brachytherapy; boost dosing to chest wall scar is usually treated with photons or electrons, the guidelines note.

The guidelines recommend weekly imaging to ensure that daily treatment setup is consistent, but caution against routine use of daily imaging.

For whole breast irradiation, the guidelines recommend either doses of 46-50 Gy divided into 23-25 fractions (conventional fractionation), or, preferably, hypofractionated radiation delivered in doses of 40-42.5 Gy over 15-16 fractions, with all doses given 5 days per week. Patients at higher risk for recurrence should receive boost doses to the tumor bed, typically at doses of 10-16 Gy delivered in 4-8 fractions.

The guidelines now favor hypofractionation based on long-term results from clinical trials conducted in Ontario, Canada, and in London.

In the Canadian trial (N Engl J Med. 2010;362:513-20), patients were treated with whole-breast irradiation at doses of 42.5 Gy in 16 fractions, with no boost dose.

In the Standardisation of Breast Radiotherapy B (START B) trial (Lancet Oncol. 14;11:1086-94), in London, patients received 40 Gy in 15 fractions without a boost dose.

Both trials showed that hypofractionation was associated with disease outcomes that were equivalent or better to those seen with conventional fractionation schedules, as well as equivalent or better cosmesis and adverse event profiles, Dr. Salerno said.

“This is for whole breast radiation; it is not to be routinely used in the post-mastectomy setting or when you’re treating regional nodes,” she said.

Accelerated partial breast irradiation

For accelerated partial breast irradiation (APBI), the guidelines recommend a hypofractionated schedule of 34 Gy divided into 10 fractions delivered twice daily for 5 days for brachytherapy, or 38.5 Gy in 10 twice daily fractions with external beam therapy to the tumor bed.

The NCCN guidelines for APBI are based on the American Society of Radiation Oncology (ASTRO) consensus statement for APBI. An updated statement is planned for 2017.

Omit radiation?

Results from the CALGB 9343 trial and other studies suggest that in selected women with lower risk for recurrence, radiation can be omitted without compromising survival. These include women aged 70 or older with small primary breast cancers, negative nodes, negative surgical margins, and hormone-receptor-positive disease who are treated with breast conserving surgery and go on to tamoxifen maintenance.

Post-mastectomy radiation

Following total mastectomy with surgical axillary staging, with our without reconstruction, the guidelines recommend specific steps based on the number of nodes involved and the tumor size and/or margin status.

Thus, women with four or more positive axillary nodes should receive radiation to the chest wall and infraclavicular region, supraclavicular area, internal mammary nodes, and any at-risk part of the axillary bed. If adjuvant chemotherapy is prescribed, radiation usually is given following chemotherapy completion.

For women with one to three positive axillary nodes, the guidelines urge clinicians to “strongly consider” treating them as they would women with four or more positive nodes. The same advice applies to women with negative nodes but tumors larger than 5 cm or positive surgical margins as defined by ink on tumor.

For patients with negative nodes, tumors 5 cm or smaller and negative but narrow margins (less than 1 mm), the guidelines recommend consideration of radiation to the chest wall. Patients with small tumors, no nodal involvement, and margins 1 mm or greater can be spared radiation therapy.

 

 

Recurrence

For women with local-only recurrence, the guidelines recommend mastectomy with axillary lymph node staging for those with recurrences after initial lumpectomy and radiation, or if the initial treatment was mastectomy with no radiation, surgical resection with radiation, or surgical resection alone for those women previously treated with mastectomy and radiation.

For regional or locoregional recurrences, the guidelines recommend that axillary recurrences be treated with surgery and radiation if possible, and that supraclavicular or internal mammary node recurrences be treated with radiation therapy, if possible.

HOLLYWOOD, FLA. – No huge surprises here, but newly revised National Comprehensive Cancer Network guidelines for locoregional therapy of breast cancer help clarify the optimal use of adjuvant radiation.

For example, updates to the pages on principles of radiation therapy in invasive breast cancer contain new information on optimal dosing, fractionation, treatment planning, and techniques for minimizing radiation of surrounding normal tissues, noted Dr. Kilian E. Salerno, director of breast radiation and soft tissue/melanoma radiation at the Roswell Park Cancer Institute in Buffalo, New York.

Dr. Kilian E. Salerno

“There are a number of treatment options, many more now than ever before,” she said at the annual conference of the National Comprehensive Cancer Network.

The guidelines emphasize the importance of individualized radiation therapy planning and delivery. A CT-based treatment plan can help clinicians choose target volumes and identify adjacent organs and tissues that may be at risk for radiation spill.

Recommended options for reducing radiation doses to adjacent tissues include deep breath holds and prone positioning so that the treated breast hangs down, helping to isolate it from the heart and lungs.

Whole breast irradiation

Whole breast irradiation is the standard for radiation therapy of early breast cancer following breast-conserving surgery, although partial breast irradiation, typically of lumpectomy cavities, is also commonly performed, Dr. Salerno said.

Boost radiation in conjunction with breast conserving therapy can be delivered via en face electron fields, photons, or brachytherapy; boost dosing to chest wall scar is usually treated with photons or electrons, the guidelines note.

The guidelines recommend weekly imaging to ensure that daily treatment setup is consistent, but caution against routine use of daily imaging.

For whole breast irradiation, the guidelines recommend either doses of 46-50 Gy divided into 23-25 fractions (conventional fractionation), or, preferably, hypofractionated radiation delivered in doses of 40-42.5 Gy over 15-16 fractions, with all doses given 5 days per week. Patients at higher risk for recurrence should receive boost doses to the tumor bed, typically at doses of 10-16 Gy delivered in 4-8 fractions.

The guidelines now favor hypofractionation based on long-term results from clinical trials conducted in Ontario, Canada, and in London.

In the Canadian trial (N Engl J Med. 2010;362:513-20), patients were treated with whole-breast irradiation at doses of 42.5 Gy in 16 fractions, with no boost dose.

In the Standardisation of Breast Radiotherapy B (START B) trial (Lancet Oncol. 14;11:1086-94), in London, patients received 40 Gy in 15 fractions without a boost dose.

Both trials showed that hypofractionation was associated with disease outcomes that were equivalent or better to those seen with conventional fractionation schedules, as well as equivalent or better cosmesis and adverse event profiles, Dr. Salerno said.

“This is for whole breast radiation; it is not to be routinely used in the post-mastectomy setting or when you’re treating regional nodes,” she said.

Accelerated partial breast irradiation

For accelerated partial breast irradiation (APBI), the guidelines recommend a hypofractionated schedule of 34 Gy divided into 10 fractions delivered twice daily for 5 days for brachytherapy, or 38.5 Gy in 10 twice daily fractions with external beam therapy to the tumor bed.

The NCCN guidelines for APBI are based on the American Society of Radiation Oncology (ASTRO) consensus statement for APBI. An updated statement is planned for 2017.

Omit radiation?

Results from the CALGB 9343 trial and other studies suggest that in selected women with lower risk for recurrence, radiation can be omitted without compromising survival. These include women aged 70 or older with small primary breast cancers, negative nodes, negative surgical margins, and hormone-receptor-positive disease who are treated with breast conserving surgery and go on to tamoxifen maintenance.

Post-mastectomy radiation

Following total mastectomy with surgical axillary staging, with our without reconstruction, the guidelines recommend specific steps based on the number of nodes involved and the tumor size and/or margin status.

Thus, women with four or more positive axillary nodes should receive radiation to the chest wall and infraclavicular region, supraclavicular area, internal mammary nodes, and any at-risk part of the axillary bed. If adjuvant chemotherapy is prescribed, radiation usually is given following chemotherapy completion.

For women with one to three positive axillary nodes, the guidelines urge clinicians to “strongly consider” treating them as they would women with four or more positive nodes. The same advice applies to women with negative nodes but tumors larger than 5 cm or positive surgical margins as defined by ink on tumor.

For patients with negative nodes, tumors 5 cm or smaller and negative but narrow margins (less than 1 mm), the guidelines recommend consideration of radiation to the chest wall. Patients with small tumors, no nodal involvement, and margins 1 mm or greater can be spared radiation therapy.

 

 

Recurrence

For women with local-only recurrence, the guidelines recommend mastectomy with axillary lymph node staging for those with recurrences after initial lumpectomy and radiation, or if the initial treatment was mastectomy with no radiation, surgical resection with radiation, or surgical resection alone for those women previously treated with mastectomy and radiation.

For regional or locoregional recurrences, the guidelines recommend that axillary recurrences be treated with surgery and radiation if possible, and that supraclavicular or internal mammary node recurrences be treated with radiation therapy, if possible.

References

References

Publications
Publications
Topics
Article Type
Display Headline
NCCN breast cancer RT guidelines: hypofractionation preferred
Display Headline
NCCN breast cancer RT guidelines: hypofractionation preferred
Article Source

AT THE NCCN ANNUAL CONFERENCE

PURLs Copyright

Inside the Article

Vitals

Key clinical point: NCCN guidelines on radiation therapy for locoregional treatment of breast cancer emphasize hypofractionated whole-breast irradiation for treatment of early disease.

Major finding: In clinical trials, hypofractionated whole-breast irradiation was equivalent to or better than conventional fractionation for disease control, cosmesis, and adverse events.

Data source: Review of updated NCCN guidelines and supporting evidence.

Disclosures: Dr. Salerno reported having no relevant disclosures.

NCCN fine-tunes breast cancer guidelines

Article Type
Changed
Wed, 01/04/2023 - 16:49
Display Headline
NCCN fine-tunes breast cancer guidelines

HOLLYWOOD, FLA. – The latest updates to the National Comprehensive Cancer Network guidelines on breast cancer reflect a more nuanced approach to the use of systemic therapy in both the pre- and postoperative settings, with increasing emphasis on tailoring therapies to the unique clinical profiles of individual patients, according to Dr. William Gradishar.

The revised guidelines include new recommendations about preoperative endocrine and HER2-directed therapies, optimizing adjuvant endocrine therapy in both pre- and postmenopausal women, targeted agents as partner drugs for endocrine therapy in women with metastatic breast cancer, and recommendations about preservation of fertility in younger patients, said Dr. Gradishar, chair of the Robert H. Lurie Comprehensive Cancer Center at Northwestern University in Chicago.

Dr. William J. Gradishar

“There have been some tweaks but I wouldn’t say major, major changes within the guidelines,” he said at the annual conference of the National Comprehensive Cancer Network.

Neoadjuvant therapy

One of the larger changes to this year’s guidelines is the addition of a page devoted to the principles of preoperative systemic therapy. The guidelines note that randomized clinical trials have shown similar long-term outcomes when patients receive the same treatment in either the pre- or post-op setting.

They add, however, that neoadjuvant systemic therapy “can render surgically inoperable tumors operable and offers potential benefits for patients with operable breast cancer. Importantly, preoperative systemic therapy can improve rates of breast conservation therapy eligibility and provides an opportunity to observe clinical and pathologic response to systemic therapy in an individual patient.”

The guidelines also note that a pathologic complete response to preoperative systemic therapy is predictive of “extremely favorable” disease-free and overall survival.

Patients who may be good candidates for neoadjuvant endocrine therapy include those with estrogen receptor–rich tumors (Allred score, 7 or 8), older postmenopausal women, and younger, premenopausal women with significant comorbidities that may make them poor candidates for chemotherapy, Dr. Gradishar said.

Adjuvant therapy

Regarding adjuvant therapy, the guidelines reflect data from recent clinical trials showing the benefits of both extended tamoxifen therapy, and for women who are premenopausal at diagnosis and are at high risk of recurrence, an aromatase inhibitor (AI) or steroidal aromatase inactivator plus ovarian suppression or ablation.

The recommendation to consider an AI and ovarian suppression in this population is based on data from the SOFT and TEXT trials (N Engl J Med. 2014; 371:107-18), which showed a significant reduction in the recurrence of hormone receptor–positive early breast cancer in premenopausal women treated with exemestane (Aromasin) and ovarian suppression, compared with tamoxifen and ovarian suppression.

Although data supporting the use of endocrine therapy and ovarian suppression are compelling, “we have to individualize our choice of which patient is appropriate for this, because there is a price to pay in terms of side effects, and we have to figure out if that balance is right for an individual patient,” Dr. Gradishar commented.

Anti-HER2 therapy

The guidelines state that patients with HER2-positive tumors should receive neoadjuvant systemic therapy with trastuzumab (Herceptin) for at least 9 weeks prior to surgery. For patients with stage T2 or greater or nodal status N1 or greater HER2-positive disease, a pertuzumab (Perjeta)-containing regimen may be considered.

Recurrent/Stage IV disease

Premenopausal women with recurrent or stage IV hormone receptor–positive disease should have ovarian ablation or suppression, and then be treated with a regimen for postmenopausal women, the guidelines say. Agents used in therapy for advanced disease have included a nonsteroidal or steroidal AI, a combination of exmestane and everolimus, palbociclib (Ibrance) plus letrozole (Femara), palbociclib plus fulvestrant, tamoxifen, or toremafine, megesterol acetate fluoxymesterone, or ethinyl estradiol.

The updated guidelines note that “a single study (S0226) in women with hormone receptor–positive breast cancer and no prior chemotherapy, biological therapy, or endocrine therapy for metastatic disease demonstrated that the addition of fulvestrant to anastrozole resulted in prolongation of time to progression. Subset analysis suggested that patients without prior adjuvant tamoxifen and more than 10 years since diagnosis experienced the greatest benefit. Two studies with similar design (FACT and SOFEA) demonstrated no advantage in time to progression with the addition of fulvestrant to anastrozole.”

Chemotherapy regimens for recurrent/metastatic breast cancer include various combination regimens, as well as single-agent therapy with anthracyclines, taxanes, antimetabolites, or microtubule inhibitors (all of the above preferred single agents), or with cyclophosphamide, carboplatin, docetaxel, albumin-bound paclitaxel, cisplatin, epirubicin, or ixabepilone.

For HER2-positive disease, preferred agents include pertuzumab and trastuzumab with either docetaxel (category 1 recommendation) or paclitaxel. Trastuzumab can also be combined with other agents such as paclitaxel with or without cisplatin, docetaxel, vinorelbine, or capecitabine.

 

 

For patients with recurrent HER2-positve disease with prior trastuzumab exposure, combinations may include lapatinin with capecitabine, or trastuzumab with capecitabine, lapatinib (without cytoxic therapy), or other agents.

Fertility preservation

Lastly, the revised guidelines note that all premenopausal patients should be informed about the potential effects of chemotherapy on fertility, with women who express the wish to have future pregnancies referred to fertility specialists. The options presented should be “based on patient specifics, disease stage, and biology (which determine the urgency and type and sequence of treatment). Timing and duration allowed for fertility preservation, options inclusive of oocyte and embryo cryopreservation as well as evolving technologies, and the probability of successful pregnancies subsequent to completion of breast cancer therapy are also to be discussed,” the guidelines say.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

HOLLYWOOD, FLA. – The latest updates to the National Comprehensive Cancer Network guidelines on breast cancer reflect a more nuanced approach to the use of systemic therapy in both the pre- and postoperative settings, with increasing emphasis on tailoring therapies to the unique clinical profiles of individual patients, according to Dr. William Gradishar.

The revised guidelines include new recommendations about preoperative endocrine and HER2-directed therapies, optimizing adjuvant endocrine therapy in both pre- and postmenopausal women, targeted agents as partner drugs for endocrine therapy in women with metastatic breast cancer, and recommendations about preservation of fertility in younger patients, said Dr. Gradishar, chair of the Robert H. Lurie Comprehensive Cancer Center at Northwestern University in Chicago.

Dr. William J. Gradishar

“There have been some tweaks but I wouldn’t say major, major changes within the guidelines,” he said at the annual conference of the National Comprehensive Cancer Network.

Neoadjuvant therapy

One of the larger changes to this year’s guidelines is the addition of a page devoted to the principles of preoperative systemic therapy. The guidelines note that randomized clinical trials have shown similar long-term outcomes when patients receive the same treatment in either the pre- or post-op setting.

They add, however, that neoadjuvant systemic therapy “can render surgically inoperable tumors operable and offers potential benefits for patients with operable breast cancer. Importantly, preoperative systemic therapy can improve rates of breast conservation therapy eligibility and provides an opportunity to observe clinical and pathologic response to systemic therapy in an individual patient.”

The guidelines also note that a pathologic complete response to preoperative systemic therapy is predictive of “extremely favorable” disease-free and overall survival.

Patients who may be good candidates for neoadjuvant endocrine therapy include those with estrogen receptor–rich tumors (Allred score, 7 or 8), older postmenopausal women, and younger, premenopausal women with significant comorbidities that may make them poor candidates for chemotherapy, Dr. Gradishar said.

Adjuvant therapy

Regarding adjuvant therapy, the guidelines reflect data from recent clinical trials showing the benefits of both extended tamoxifen therapy, and for women who are premenopausal at diagnosis and are at high risk of recurrence, an aromatase inhibitor (AI) or steroidal aromatase inactivator plus ovarian suppression or ablation.

The recommendation to consider an AI and ovarian suppression in this population is based on data from the SOFT and TEXT trials (N Engl J Med. 2014; 371:107-18), which showed a significant reduction in the recurrence of hormone receptor–positive early breast cancer in premenopausal women treated with exemestane (Aromasin) and ovarian suppression, compared with tamoxifen and ovarian suppression.

Although data supporting the use of endocrine therapy and ovarian suppression are compelling, “we have to individualize our choice of which patient is appropriate for this, because there is a price to pay in terms of side effects, and we have to figure out if that balance is right for an individual patient,” Dr. Gradishar commented.

Anti-HER2 therapy

The guidelines state that patients with HER2-positive tumors should receive neoadjuvant systemic therapy with trastuzumab (Herceptin) for at least 9 weeks prior to surgery. For patients with stage T2 or greater or nodal status N1 or greater HER2-positive disease, a pertuzumab (Perjeta)-containing regimen may be considered.

Recurrent/Stage IV disease

Premenopausal women with recurrent or stage IV hormone receptor–positive disease should have ovarian ablation or suppression, and then be treated with a regimen for postmenopausal women, the guidelines say. Agents used in therapy for advanced disease have included a nonsteroidal or steroidal AI, a combination of exmestane and everolimus, palbociclib (Ibrance) plus letrozole (Femara), palbociclib plus fulvestrant, tamoxifen, or toremafine, megesterol acetate fluoxymesterone, or ethinyl estradiol.

The updated guidelines note that “a single study (S0226) in women with hormone receptor–positive breast cancer and no prior chemotherapy, biological therapy, or endocrine therapy for metastatic disease demonstrated that the addition of fulvestrant to anastrozole resulted in prolongation of time to progression. Subset analysis suggested that patients without prior adjuvant tamoxifen and more than 10 years since diagnosis experienced the greatest benefit. Two studies with similar design (FACT and SOFEA) demonstrated no advantage in time to progression with the addition of fulvestrant to anastrozole.”

Chemotherapy regimens for recurrent/metastatic breast cancer include various combination regimens, as well as single-agent therapy with anthracyclines, taxanes, antimetabolites, or microtubule inhibitors (all of the above preferred single agents), or with cyclophosphamide, carboplatin, docetaxel, albumin-bound paclitaxel, cisplatin, epirubicin, or ixabepilone.

For HER2-positive disease, preferred agents include pertuzumab and trastuzumab with either docetaxel (category 1 recommendation) or paclitaxel. Trastuzumab can also be combined with other agents such as paclitaxel with or without cisplatin, docetaxel, vinorelbine, or capecitabine.

 

 

For patients with recurrent HER2-positve disease with prior trastuzumab exposure, combinations may include lapatinin with capecitabine, or trastuzumab with capecitabine, lapatinib (without cytoxic therapy), or other agents.

Fertility preservation

Lastly, the revised guidelines note that all premenopausal patients should be informed about the potential effects of chemotherapy on fertility, with women who express the wish to have future pregnancies referred to fertility specialists. The options presented should be “based on patient specifics, disease stage, and biology (which determine the urgency and type and sequence of treatment). Timing and duration allowed for fertility preservation, options inclusive of oocyte and embryo cryopreservation as well as evolving technologies, and the probability of successful pregnancies subsequent to completion of breast cancer therapy are also to be discussed,” the guidelines say.

HOLLYWOOD, FLA. – The latest updates to the National Comprehensive Cancer Network guidelines on breast cancer reflect a more nuanced approach to the use of systemic therapy in both the pre- and postoperative settings, with increasing emphasis on tailoring therapies to the unique clinical profiles of individual patients, according to Dr. William Gradishar.

The revised guidelines include new recommendations about preoperative endocrine and HER2-directed therapies, optimizing adjuvant endocrine therapy in both pre- and postmenopausal women, targeted agents as partner drugs for endocrine therapy in women with metastatic breast cancer, and recommendations about preservation of fertility in younger patients, said Dr. Gradishar, chair of the Robert H. Lurie Comprehensive Cancer Center at Northwestern University in Chicago.

Dr. William J. Gradishar

“There have been some tweaks but I wouldn’t say major, major changes within the guidelines,” he said at the annual conference of the National Comprehensive Cancer Network.

Neoadjuvant therapy

One of the larger changes to this year’s guidelines is the addition of a page devoted to the principles of preoperative systemic therapy. The guidelines note that randomized clinical trials have shown similar long-term outcomes when patients receive the same treatment in either the pre- or post-op setting.

They add, however, that neoadjuvant systemic therapy “can render surgically inoperable tumors operable and offers potential benefits for patients with operable breast cancer. Importantly, preoperative systemic therapy can improve rates of breast conservation therapy eligibility and provides an opportunity to observe clinical and pathologic response to systemic therapy in an individual patient.”

The guidelines also note that a pathologic complete response to preoperative systemic therapy is predictive of “extremely favorable” disease-free and overall survival.

Patients who may be good candidates for neoadjuvant endocrine therapy include those with estrogen receptor–rich tumors (Allred score, 7 or 8), older postmenopausal women, and younger, premenopausal women with significant comorbidities that may make them poor candidates for chemotherapy, Dr. Gradishar said.

Adjuvant therapy

Regarding adjuvant therapy, the guidelines reflect data from recent clinical trials showing the benefits of both extended tamoxifen therapy, and for women who are premenopausal at diagnosis and are at high risk of recurrence, an aromatase inhibitor (AI) or steroidal aromatase inactivator plus ovarian suppression or ablation.

The recommendation to consider an AI and ovarian suppression in this population is based on data from the SOFT and TEXT trials (N Engl J Med. 2014; 371:107-18), which showed a significant reduction in the recurrence of hormone receptor–positive early breast cancer in premenopausal women treated with exemestane (Aromasin) and ovarian suppression, compared with tamoxifen and ovarian suppression.

Although data supporting the use of endocrine therapy and ovarian suppression are compelling, “we have to individualize our choice of which patient is appropriate for this, because there is a price to pay in terms of side effects, and we have to figure out if that balance is right for an individual patient,” Dr. Gradishar commented.

Anti-HER2 therapy

The guidelines state that patients with HER2-positive tumors should receive neoadjuvant systemic therapy with trastuzumab (Herceptin) for at least 9 weeks prior to surgery. For patients with stage T2 or greater or nodal status N1 or greater HER2-positive disease, a pertuzumab (Perjeta)-containing regimen may be considered.

Recurrent/Stage IV disease

Premenopausal women with recurrent or stage IV hormone receptor–positive disease should have ovarian ablation or suppression, and then be treated with a regimen for postmenopausal women, the guidelines say. Agents used in therapy for advanced disease have included a nonsteroidal or steroidal AI, a combination of exmestane and everolimus, palbociclib (Ibrance) plus letrozole (Femara), palbociclib plus fulvestrant, tamoxifen, or toremafine, megesterol acetate fluoxymesterone, or ethinyl estradiol.

The updated guidelines note that “a single study (S0226) in women with hormone receptor–positive breast cancer and no prior chemotherapy, biological therapy, or endocrine therapy for metastatic disease demonstrated that the addition of fulvestrant to anastrozole resulted in prolongation of time to progression. Subset analysis suggested that patients without prior adjuvant tamoxifen and more than 10 years since diagnosis experienced the greatest benefit. Two studies with similar design (FACT and SOFEA) demonstrated no advantage in time to progression with the addition of fulvestrant to anastrozole.”

Chemotherapy regimens for recurrent/metastatic breast cancer include various combination regimens, as well as single-agent therapy with anthracyclines, taxanes, antimetabolites, or microtubule inhibitors (all of the above preferred single agents), or with cyclophosphamide, carboplatin, docetaxel, albumin-bound paclitaxel, cisplatin, epirubicin, or ixabepilone.

For HER2-positive disease, preferred agents include pertuzumab and trastuzumab with either docetaxel (category 1 recommendation) or paclitaxel. Trastuzumab can also be combined with other agents such as paclitaxel with or without cisplatin, docetaxel, vinorelbine, or capecitabine.

 

 

For patients with recurrent HER2-positve disease with prior trastuzumab exposure, combinations may include lapatinin with capecitabine, or trastuzumab with capecitabine, lapatinib (without cytoxic therapy), or other agents.

Fertility preservation

Lastly, the revised guidelines note that all premenopausal patients should be informed about the potential effects of chemotherapy on fertility, with women who express the wish to have future pregnancies referred to fertility specialists. The options presented should be “based on patient specifics, disease stage, and biology (which determine the urgency and type and sequence of treatment). Timing and duration allowed for fertility preservation, options inclusive of oocyte and embryo cryopreservation as well as evolving technologies, and the probability of successful pregnancies subsequent to completion of breast cancer therapy are also to be discussed,” the guidelines say.

References

References

Publications
Publications
Topics
Article Type
Display Headline
NCCN fine-tunes breast cancer guidelines
Display Headline
NCCN fine-tunes breast cancer guidelines
Sections
Article Source

AT THE NCCN ANNUAL CONFERENCE

PURLs Copyright

Inside the Article

Inking bests suturing to mark breast tumor margins

Article Type
Changed
Wed, 01/04/2023 - 16:49
Display Headline
Inking bests suturing to mark breast tumor margins

BOSTON – With art supplies and “mystery” sutures, a team of investigators has determined that immediate intraoperative inking of lumpectomy specimens appears to be a better method than suture placement for orienting specimens for pathology, a finding that could reduce re-excisions.

“Intraoperative specimen suturing appears to be inaccurate for margin identification, and additional means by the surgeon to improve the accuracy are needed. This could be either immediate inking or routine excision of shave margins, either at the original surgery or when you go back for re-excision, to take all the margins instead of one specific margin,” Dr. Angel Arnaout, a breast surgeon at the University of Ottawa, Ont., said at the annual Society of Surgical Oncology Cancer Symposium.

She presented results of the randomized, blinded SMART (Specimen Margin Assessment Technique) trial comparing suture placement with intraoperative inking within the same surgical specimen.

Positive resection margins during breast-conserving surgery are associated with a twofold increase in the risk of ipsilateral tumor recurrence, and require re-excision or conversion to mastectomy. Re-excisions rates range from 25% to 45%, but in the majority of cases – 53% to 65% – no additional disease is found, Dr. Arnaout said.

Dr. Angel Arnaout

In addition to adding to patient distress, re-excisions diminish cosmetic results because they take additional tissue, and delay the start of adjuvant breast cancer therapy, she noted.

Surgeons typically orient specimens for pathologists by placing two or three sutures in the center of the margin surfaces or, less frequently, by intraoperative marking of the margins with ink. But in transit from the OR to the pathology lab, specimens often flatten or “pancake” under their own weight, making it challenging for the pathologist to identify the originally marked margins.

To test whether suturing or inking is better at identifying the extent of tumors during breast-conserving surgery, the investigators devised a strategy using benign breast tissue removed during prophylactic mastectomy or reduction mammoplasty.

The surgeons first performed a sham lumpectomy within the tissue. They then painted the specimen margins in the OR using a phospholuminescent paint obtained from an art supply store. The paint dries clear and colorless, but glows in different colors under black light. By using the paint, carefully selected so as not to interfere with pathology staining, the investigators were able to blind the pathologists to the actual orientation of the margins.

While the tissue was still in the OR, the surgeons then placed two or three orienting sutures in customary locations, plus a third, “mystery” suture at a location known only to the surgeon.

In the pathology lab, the assistant was asked to look at the sutures and outline in black ink the edges of the margins using the sutures for guidance.

The specimen was then examined under black light to determine the degree of discordance between surgeons and pathologist for identification of margins using the suture and painting methods (primary outcome), and to evaluate the discrepancy in the extent of margin surface areas as identified by the surgeons and the pathologists (secondary outcome).

“We asked breast surgeons ‘what would be a clinically significant discordance rate for you?’ and most of them said between 10% and 20%, so we looked for a 15% discordance rate as being clinically significant between the two margin assessment techniques,” Dr. Arnaout said.

They found that the overall discordance in the location of the mystery suture between surgeons and pathologists was similar whether the samples contained two or three location sutures, with discordance rates of 46% (34 of 75 samples) for two sutures, and 47% (41 of 88 samples) for three sutures.

For the secondary outcome of discordance in the extent of margins, they found that pathologists tended to estimate the mean surface of the anterior and posterior margins to be significantly larger than the surgeons did (P less than .01), while significantly underestimating the superior, inferior, and lateral margins (P less than .01 for superior and inferior, and P = .04 for lateral margins).

Examination under black light showed that often two or three additional margins would be included by the surgeon within what the pathologist had identified as the anterior margin.

“This has implications for the surgeon and for the patient,” Dr. Arnaout said. “For most of us that do lumpectomies that go from skin down to chest wall, if an anterior margin or posterior margin is positive, a lot of times we would fight not to go back because we would say there is no further room for re-excision.”

But if the tumor is within what is actually the superior margin but labeled by the pathologist as an anterior margin, the patient may miss an opportunity for successful re-excision, she explained.

 

 

The study was limited by the fact that the sham lumpectomy specimens did not contain skin or muscle, which would have allowed for more accurate margin orientation in an actual operative setting; by lack of compression of specimens with small nonpalpable lesions in containers, which further distorts specimens; and by the use of smaller lumpectomy specimens than normally obtained during cancer surgery, which could have resulted in overestimation of the discordance that might occur when larger specimens are taken, Dr. Arnaout said.

“The conclusion of our study is that specimen margin orientation really should be defined by the surgeon who knows the original shape and orientation of the tissue during surgery, and not to rely on the pathology to reorient based on some sutures placed in the centers of the specimens without defining the extent of the surface area of each of the margins,” she said.

The Canadian Cancer Society Research Institute and the Canadian Surgical Research Fund supported the study. Dr. Arnaout and coauthors reported no conflicts of interest.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

BOSTON – With art supplies and “mystery” sutures, a team of investigators has determined that immediate intraoperative inking of lumpectomy specimens appears to be a better method than suture placement for orienting specimens for pathology, a finding that could reduce re-excisions.

“Intraoperative specimen suturing appears to be inaccurate for margin identification, and additional means by the surgeon to improve the accuracy are needed. This could be either immediate inking or routine excision of shave margins, either at the original surgery or when you go back for re-excision, to take all the margins instead of one specific margin,” Dr. Angel Arnaout, a breast surgeon at the University of Ottawa, Ont., said at the annual Society of Surgical Oncology Cancer Symposium.

She presented results of the randomized, blinded SMART (Specimen Margin Assessment Technique) trial comparing suture placement with intraoperative inking within the same surgical specimen.

Positive resection margins during breast-conserving surgery are associated with a twofold increase in the risk of ipsilateral tumor recurrence, and require re-excision or conversion to mastectomy. Re-excisions rates range from 25% to 45%, but in the majority of cases – 53% to 65% – no additional disease is found, Dr. Arnaout said.

Dr. Angel Arnaout

In addition to adding to patient distress, re-excisions diminish cosmetic results because they take additional tissue, and delay the start of adjuvant breast cancer therapy, she noted.

Surgeons typically orient specimens for pathologists by placing two or three sutures in the center of the margin surfaces or, less frequently, by intraoperative marking of the margins with ink. But in transit from the OR to the pathology lab, specimens often flatten or “pancake” under their own weight, making it challenging for the pathologist to identify the originally marked margins.

To test whether suturing or inking is better at identifying the extent of tumors during breast-conserving surgery, the investigators devised a strategy using benign breast tissue removed during prophylactic mastectomy or reduction mammoplasty.

The surgeons first performed a sham lumpectomy within the tissue. They then painted the specimen margins in the OR using a phospholuminescent paint obtained from an art supply store. The paint dries clear and colorless, but glows in different colors under black light. By using the paint, carefully selected so as not to interfere with pathology staining, the investigators were able to blind the pathologists to the actual orientation of the margins.

While the tissue was still in the OR, the surgeons then placed two or three orienting sutures in customary locations, plus a third, “mystery” suture at a location known only to the surgeon.

In the pathology lab, the assistant was asked to look at the sutures and outline in black ink the edges of the margins using the sutures for guidance.

The specimen was then examined under black light to determine the degree of discordance between surgeons and pathologist for identification of margins using the suture and painting methods (primary outcome), and to evaluate the discrepancy in the extent of margin surface areas as identified by the surgeons and the pathologists (secondary outcome).

“We asked breast surgeons ‘what would be a clinically significant discordance rate for you?’ and most of them said between 10% and 20%, so we looked for a 15% discordance rate as being clinically significant between the two margin assessment techniques,” Dr. Arnaout said.

They found that the overall discordance in the location of the mystery suture between surgeons and pathologists was similar whether the samples contained two or three location sutures, with discordance rates of 46% (34 of 75 samples) for two sutures, and 47% (41 of 88 samples) for three sutures.

For the secondary outcome of discordance in the extent of margins, they found that pathologists tended to estimate the mean surface of the anterior and posterior margins to be significantly larger than the surgeons did (P less than .01), while significantly underestimating the superior, inferior, and lateral margins (P less than .01 for superior and inferior, and P = .04 for lateral margins).

Examination under black light showed that often two or three additional margins would be included by the surgeon within what the pathologist had identified as the anterior margin.

“This has implications for the surgeon and for the patient,” Dr. Arnaout said. “For most of us that do lumpectomies that go from skin down to chest wall, if an anterior margin or posterior margin is positive, a lot of times we would fight not to go back because we would say there is no further room for re-excision.”

But if the tumor is within what is actually the superior margin but labeled by the pathologist as an anterior margin, the patient may miss an opportunity for successful re-excision, she explained.

 

 

The study was limited by the fact that the sham lumpectomy specimens did not contain skin or muscle, which would have allowed for more accurate margin orientation in an actual operative setting; by lack of compression of specimens with small nonpalpable lesions in containers, which further distorts specimens; and by the use of smaller lumpectomy specimens than normally obtained during cancer surgery, which could have resulted in overestimation of the discordance that might occur when larger specimens are taken, Dr. Arnaout said.

“The conclusion of our study is that specimen margin orientation really should be defined by the surgeon who knows the original shape and orientation of the tissue during surgery, and not to rely on the pathology to reorient based on some sutures placed in the centers of the specimens without defining the extent of the surface area of each of the margins,” she said.

The Canadian Cancer Society Research Institute and the Canadian Surgical Research Fund supported the study. Dr. Arnaout and coauthors reported no conflicts of interest.

BOSTON – With art supplies and “mystery” sutures, a team of investigators has determined that immediate intraoperative inking of lumpectomy specimens appears to be a better method than suture placement for orienting specimens for pathology, a finding that could reduce re-excisions.

“Intraoperative specimen suturing appears to be inaccurate for margin identification, and additional means by the surgeon to improve the accuracy are needed. This could be either immediate inking or routine excision of shave margins, either at the original surgery or when you go back for re-excision, to take all the margins instead of one specific margin,” Dr. Angel Arnaout, a breast surgeon at the University of Ottawa, Ont., said at the annual Society of Surgical Oncology Cancer Symposium.

She presented results of the randomized, blinded SMART (Specimen Margin Assessment Technique) trial comparing suture placement with intraoperative inking within the same surgical specimen.

Positive resection margins during breast-conserving surgery are associated with a twofold increase in the risk of ipsilateral tumor recurrence, and require re-excision or conversion to mastectomy. Re-excisions rates range from 25% to 45%, but in the majority of cases – 53% to 65% – no additional disease is found, Dr. Arnaout said.

Dr. Angel Arnaout

In addition to adding to patient distress, re-excisions diminish cosmetic results because they take additional tissue, and delay the start of adjuvant breast cancer therapy, she noted.

Surgeons typically orient specimens for pathologists by placing two or three sutures in the center of the margin surfaces or, less frequently, by intraoperative marking of the margins with ink. But in transit from the OR to the pathology lab, specimens often flatten or “pancake” under their own weight, making it challenging for the pathologist to identify the originally marked margins.

To test whether suturing or inking is better at identifying the extent of tumors during breast-conserving surgery, the investigators devised a strategy using benign breast tissue removed during prophylactic mastectomy or reduction mammoplasty.

The surgeons first performed a sham lumpectomy within the tissue. They then painted the specimen margins in the OR using a phospholuminescent paint obtained from an art supply store. The paint dries clear and colorless, but glows in different colors under black light. By using the paint, carefully selected so as not to interfere with pathology staining, the investigators were able to blind the pathologists to the actual orientation of the margins.

While the tissue was still in the OR, the surgeons then placed two or three orienting sutures in customary locations, plus a third, “mystery” suture at a location known only to the surgeon.

In the pathology lab, the assistant was asked to look at the sutures and outline in black ink the edges of the margins using the sutures for guidance.

The specimen was then examined under black light to determine the degree of discordance between surgeons and pathologist for identification of margins using the suture and painting methods (primary outcome), and to evaluate the discrepancy in the extent of margin surface areas as identified by the surgeons and the pathologists (secondary outcome).

“We asked breast surgeons ‘what would be a clinically significant discordance rate for you?’ and most of them said between 10% and 20%, so we looked for a 15% discordance rate as being clinically significant between the two margin assessment techniques,” Dr. Arnaout said.

They found that the overall discordance in the location of the mystery suture between surgeons and pathologists was similar whether the samples contained two or three location sutures, with discordance rates of 46% (34 of 75 samples) for two sutures, and 47% (41 of 88 samples) for three sutures.

For the secondary outcome of discordance in the extent of margins, they found that pathologists tended to estimate the mean surface of the anterior and posterior margins to be significantly larger than the surgeons did (P less than .01), while significantly underestimating the superior, inferior, and lateral margins (P less than .01 for superior and inferior, and P = .04 for lateral margins).

Examination under black light showed that often two or three additional margins would be included by the surgeon within what the pathologist had identified as the anterior margin.

“This has implications for the surgeon and for the patient,” Dr. Arnaout said. “For most of us that do lumpectomies that go from skin down to chest wall, if an anterior margin or posterior margin is positive, a lot of times we would fight not to go back because we would say there is no further room for re-excision.”

But if the tumor is within what is actually the superior margin but labeled by the pathologist as an anterior margin, the patient may miss an opportunity for successful re-excision, she explained.

 

 

The study was limited by the fact that the sham lumpectomy specimens did not contain skin or muscle, which would have allowed for more accurate margin orientation in an actual operative setting; by lack of compression of specimens with small nonpalpable lesions in containers, which further distorts specimens; and by the use of smaller lumpectomy specimens than normally obtained during cancer surgery, which could have resulted in overestimation of the discordance that might occur when larger specimens are taken, Dr. Arnaout said.

“The conclusion of our study is that specimen margin orientation really should be defined by the surgeon who knows the original shape and orientation of the tissue during surgery, and not to rely on the pathology to reorient based on some sutures placed in the centers of the specimens without defining the extent of the surface area of each of the margins,” she said.

The Canadian Cancer Society Research Institute and the Canadian Surgical Research Fund supported the study. Dr. Arnaout and coauthors reported no conflicts of interest.

References

References

Publications
Publications
Topics
Article Type
Display Headline
Inking bests suturing to mark breast tumor margins
Display Headline
Inking bests suturing to mark breast tumor margins
Sections
Article Source

FROM SSO 2016

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Intraoperative suture placement is an inaccurate method for orienting breast tumor specimens for pathology.

Major finding: Discordance in identifying tumor margin surface between surgeons and pathologists was similar whether the samples contained two or three location sutures, with discordance rates of 46% (34 of 75 samples) for two sutures, and 47% (41 of 88 samples) for three sutures.

Data source: Randomized clinical trial of 163 specimens obtained from 49 patients.

Disclosures: The Canadian Cancer Society Research Institute and the Canadian Surgical Research Fund supported the study. Dr. Arnaout and coauthors reported no conflicts of interest.

Better sarcoma outcomes at high-volume centers

Article Type
Changed
Fri, 01/04/2019 - 13:13
Display Headline
Better sarcoma outcomes at high-volume centers

BOSTON – In sarcoma as in other cancers, experience counts.

That’s the conclusion of investigators who found that patients with extra-abdominal sarcomas who were treated in high-volume hospitals had half the 30-day mortality rate, higher likelihood of negative surgical margins, and better overall survival, compared with patients treated in low-volume hospitals.

“It appears there is a direct association between hospital volume and short-term as well as long-term outcomes for soft-tissue sarcomas outside the abdomen,” said Dr. Sanjay P. Bagaria of the Mayo Clinic in Jacksonville, Fla.

The findings support centralization of services at the national level in centers specializing in the management of sarcomas, he said at the annual Society of Surgical Oncology Cancer Symposium.

Previous studies have shown that patients treated in high-volume centers for cancers of the esophagus, pancreas, and lung have better outcomes than patients treated in low-volume centers, he noted.

Given the rarity of sarcomas, with an incidence of approximately 12,000 in the U.S. annually, their complexity, with more than 60 histologic subtypes, and the multimodality approach required for them, it seemed likely that a positive association between volume and outcomes could be found, Dr. Bagaria said.

He and colleagues queried the U.S. National Cancer Database, a hospital registry of data from more than 1,500 facilities accredited by the Commission on Cancer.

They drew records on all patients diagnosed with extra-abdominal sarcomas from 2003 through 2007 who underwent surgery at the reporting hospitals, and divided the cases into terciles as either low volume (3 or fewer surgical cases per year), medium volume (3.2-11.6 per year), or high volume (12 or more cases per year).

One third (33%) of all cases were concentrated in just 44 high-volume hospitals, which comprised just 4% of the total hospital sample of 1,163. An additional third (34%) of cases were managed among 196 medium-volume hospitals (17% of the hospital sample), and the remaining third (33%) were spread among 923 low-volume hospitals (79%).

The 30-day mortality rates for low-, medium-, and high-volume hospitals, respectively, were 1.7%, 1.1%, and 0.6% (P less than .0001).

Similarly, the rates of negative margins (R0 resections) were 73.%, 78.2%, and 84.2% (P less than .0001).

Five-year overall survival was identical for low- and medium-volume centers (65% each), but was significantly better for patients treated at high-volume centers (69%, P less than .001)

Compared with low-volume centers, patients treated at high-volume centers had an adjusted odds ratio (OR) for 30-day mortality of 0.46 (P = .01), an adjusted OR for R0 margins of 1.87 (P less than .001), and OR for overall mortality of 0.92 (P = .04).

Dr. Bagaria noted that the study was limited by missing data about disease-specific survival and by possible selection bias associated with the choice of Commission on Cancer-accredited institutions, which account for 70% of cancer cases nationwide but comprise one-third of all hospitals.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

BOSTON – In sarcoma as in other cancers, experience counts.

That’s the conclusion of investigators who found that patients with extra-abdominal sarcomas who were treated in high-volume hospitals had half the 30-day mortality rate, higher likelihood of negative surgical margins, and better overall survival, compared with patients treated in low-volume hospitals.

“It appears there is a direct association between hospital volume and short-term as well as long-term outcomes for soft-tissue sarcomas outside the abdomen,” said Dr. Sanjay P. Bagaria of the Mayo Clinic in Jacksonville, Fla.

The findings support centralization of services at the national level in centers specializing in the management of sarcomas, he said at the annual Society of Surgical Oncology Cancer Symposium.

Previous studies have shown that patients treated in high-volume centers for cancers of the esophagus, pancreas, and lung have better outcomes than patients treated in low-volume centers, he noted.

Given the rarity of sarcomas, with an incidence of approximately 12,000 in the U.S. annually, their complexity, with more than 60 histologic subtypes, and the multimodality approach required for them, it seemed likely that a positive association between volume and outcomes could be found, Dr. Bagaria said.

He and colleagues queried the U.S. National Cancer Database, a hospital registry of data from more than 1,500 facilities accredited by the Commission on Cancer.

They drew records on all patients diagnosed with extra-abdominal sarcomas from 2003 through 2007 who underwent surgery at the reporting hospitals, and divided the cases into terciles as either low volume (3 or fewer surgical cases per year), medium volume (3.2-11.6 per year), or high volume (12 or more cases per year).

One third (33%) of all cases were concentrated in just 44 high-volume hospitals, which comprised just 4% of the total hospital sample of 1,163. An additional third (34%) of cases were managed among 196 medium-volume hospitals (17% of the hospital sample), and the remaining third (33%) were spread among 923 low-volume hospitals (79%).

The 30-day mortality rates for low-, medium-, and high-volume hospitals, respectively, were 1.7%, 1.1%, and 0.6% (P less than .0001).

Similarly, the rates of negative margins (R0 resections) were 73.%, 78.2%, and 84.2% (P less than .0001).

Five-year overall survival was identical for low- and medium-volume centers (65% each), but was significantly better for patients treated at high-volume centers (69%, P less than .001)

Compared with low-volume centers, patients treated at high-volume centers had an adjusted odds ratio (OR) for 30-day mortality of 0.46 (P = .01), an adjusted OR for R0 margins of 1.87 (P less than .001), and OR for overall mortality of 0.92 (P = .04).

Dr. Bagaria noted that the study was limited by missing data about disease-specific survival and by possible selection bias associated with the choice of Commission on Cancer-accredited institutions, which account for 70% of cancer cases nationwide but comprise one-third of all hospitals.

BOSTON – In sarcoma as in other cancers, experience counts.

That’s the conclusion of investigators who found that patients with extra-abdominal sarcomas who were treated in high-volume hospitals had half the 30-day mortality rate, higher likelihood of negative surgical margins, and better overall survival, compared with patients treated in low-volume hospitals.

“It appears there is a direct association between hospital volume and short-term as well as long-term outcomes for soft-tissue sarcomas outside the abdomen,” said Dr. Sanjay P. Bagaria of the Mayo Clinic in Jacksonville, Fla.

The findings support centralization of services at the national level in centers specializing in the management of sarcomas, he said at the annual Society of Surgical Oncology Cancer Symposium.

Previous studies have shown that patients treated in high-volume centers for cancers of the esophagus, pancreas, and lung have better outcomes than patients treated in low-volume centers, he noted.

Given the rarity of sarcomas, with an incidence of approximately 12,000 in the U.S. annually, their complexity, with more than 60 histologic subtypes, and the multimodality approach required for them, it seemed likely that a positive association between volume and outcomes could be found, Dr. Bagaria said.

He and colleagues queried the U.S. National Cancer Database, a hospital registry of data from more than 1,500 facilities accredited by the Commission on Cancer.

They drew records on all patients diagnosed with extra-abdominal sarcomas from 2003 through 2007 who underwent surgery at the reporting hospitals, and divided the cases into terciles as either low volume (3 or fewer surgical cases per year), medium volume (3.2-11.6 per year), or high volume (12 or more cases per year).

One third (33%) of all cases were concentrated in just 44 high-volume hospitals, which comprised just 4% of the total hospital sample of 1,163. An additional third (34%) of cases were managed among 196 medium-volume hospitals (17% of the hospital sample), and the remaining third (33%) were spread among 923 low-volume hospitals (79%).

The 30-day mortality rates for low-, medium-, and high-volume hospitals, respectively, were 1.7%, 1.1%, and 0.6% (P less than .0001).

Similarly, the rates of negative margins (R0 resections) were 73.%, 78.2%, and 84.2% (P less than .0001).

Five-year overall survival was identical for low- and medium-volume centers (65% each), but was significantly better for patients treated at high-volume centers (69%, P less than .001)

Compared with low-volume centers, patients treated at high-volume centers had an adjusted odds ratio (OR) for 30-day mortality of 0.46 (P = .01), an adjusted OR for R0 margins of 1.87 (P less than .001), and OR for overall mortality of 0.92 (P = .04).

Dr. Bagaria noted that the study was limited by missing data about disease-specific survival and by possible selection bias associated with the choice of Commission on Cancer-accredited institutions, which account for 70% of cancer cases nationwide but comprise one-third of all hospitals.

References

References

Publications
Publications
Topics
Article Type
Display Headline
Better sarcoma outcomes at high-volume centers
Display Headline
Better sarcoma outcomes at high-volume centers
Sections
Article Source

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Surgical volume, a surrogate for experience, has been shown to have a direct correlation with patient outcomes for cancers of the esophagus, lung, and pancreas, and this appears to be true for sarcomas as well.

Major finding: Patients treated for sarcoma at high-volume centers had lower 30-day and overall mortality and a higher probability of negative margins than those treated at low-volume centers.

Data source: Retrospective review of data on 14,634 patients treated at 1,163 U.S. hospitals.

Disclosures: The authors reported no relevant disclosures.

CT of chest, extremity effective for sarcoma follow-up

Article Type
Changed
Fri, 01/04/2019 - 13:13
Display Headline
CT of chest, extremity effective for sarcoma follow-up

BOSTON – CT scans appear to be effective for detecting local recurrences and pulmonary metastases in patients treated for soft-tissue sarcomas of the extremities, for about a third less than the cost of follow-up with MRI.

In a retrospective study by Dr. Allison Maciver and her colleagues, among 91 patients with soft-tissue sarcomas of the extremity followed with CT, 11 patients had a total of 14 local recurrences detected on CT, and 11 of the recurrences were in patients who were clinically asymptomatic.

Surveillance CT also identified 15 cases of pulmonary metastases, and 4 incidental second primary malignancies, Dr. Maciver of the Roswell Park Cancer Institute in Buffalo, N.Y., and her coinvestigators found, and there was only one false-positive recurrence.

The benefits of CT over extremity MRI in this population include decreased imaging time, lower cost, and a larger field of view, allowing for detection of second primary malignancies, she noted in a poster session at the annual Society of Surgical Oncology Cancer Symposium.

Many sarcomas of the extremities are highly aggressive, and timely detection of local recurrences could improve chances for limb-sparing salvage therapies. Although MRI has typically been used to follow patients with sarcomas, it is expensive and has a limited field of view, Dr. Maciver said.

In addition, the risk of pulmonary metastases with some soft-tissue sarcomas is high, necessitating the use of chest CT as a surveillance tool.

To see whether CT scans of the chest and extremities could be a cost-effective surveillance strategy for both local recurrences and pulmonary metastases, the investigators did a retrospective study of a prospective database of patients who underwent surgical resection for soft-tissue sarcomas of the extremities from 2001 through 2014 and who had CT as the primary follow-up imaging modality.

They identified a total of 91 high-risk patients followed for a median of 50.5 months. The patients had an estimated 5-year freedom from local recurrence of 82%, and from distant recurrence of 80%. Five-year overall survival was 76%.

Of the 15 patients found on CT to have pulmonary metastases, there were 4 incidentally discovered second primary cancers, including 1 each of non–small cell lung cancer, pancreatic adenocarcinoma, Merkel cell carcinomatosis, and myxofibrosarcoma. There were no false-positive pulmonary metastases.

The estimated cost of 10 years of surveillance, based on 2014 gross technical costs, was $64,969 per patient for chest CT and extremity MRI, compared with $41,595 per patient for chest and extremity CT surveillance, a potential cost savings with the CT-only strategy of $23,374 per patient.

The investigators said that the overall benefits of CT, including the cost savings in an accountable care organization model, “appear to outweigh the slightly increased radiation exposure.”

The study was internally funded. The authors reported having no relevant financial disclosures.

References

Click for Credit Link
Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Click for Credit Link
Click for Credit Link
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

BOSTON – CT scans appear to be effective for detecting local recurrences and pulmonary metastases in patients treated for soft-tissue sarcomas of the extremities, for about a third less than the cost of follow-up with MRI.

In a retrospective study by Dr. Allison Maciver and her colleagues, among 91 patients with soft-tissue sarcomas of the extremity followed with CT, 11 patients had a total of 14 local recurrences detected on CT, and 11 of the recurrences were in patients who were clinically asymptomatic.

Surveillance CT also identified 15 cases of pulmonary metastases, and 4 incidental second primary malignancies, Dr. Maciver of the Roswell Park Cancer Institute in Buffalo, N.Y., and her coinvestigators found, and there was only one false-positive recurrence.

The benefits of CT over extremity MRI in this population include decreased imaging time, lower cost, and a larger field of view, allowing for detection of second primary malignancies, she noted in a poster session at the annual Society of Surgical Oncology Cancer Symposium.

Many sarcomas of the extremities are highly aggressive, and timely detection of local recurrences could improve chances for limb-sparing salvage therapies. Although MRI has typically been used to follow patients with sarcomas, it is expensive and has a limited field of view, Dr. Maciver said.

In addition, the risk of pulmonary metastases with some soft-tissue sarcomas is high, necessitating the use of chest CT as a surveillance tool.

To see whether CT scans of the chest and extremities could be a cost-effective surveillance strategy for both local recurrences and pulmonary metastases, the investigators did a retrospective study of a prospective database of patients who underwent surgical resection for soft-tissue sarcomas of the extremities from 2001 through 2014 and who had CT as the primary follow-up imaging modality.

They identified a total of 91 high-risk patients followed for a median of 50.5 months. The patients had an estimated 5-year freedom from local recurrence of 82%, and from distant recurrence of 80%. Five-year overall survival was 76%.

Of the 15 patients found on CT to have pulmonary metastases, there were 4 incidentally discovered second primary cancers, including 1 each of non–small cell lung cancer, pancreatic adenocarcinoma, Merkel cell carcinomatosis, and myxofibrosarcoma. There were no false-positive pulmonary metastases.

The estimated cost of 10 years of surveillance, based on 2014 gross technical costs, was $64,969 per patient for chest CT and extremity MRI, compared with $41,595 per patient for chest and extremity CT surveillance, a potential cost savings with the CT-only strategy of $23,374 per patient.

The investigators said that the overall benefits of CT, including the cost savings in an accountable care organization model, “appear to outweigh the slightly increased radiation exposure.”

The study was internally funded. The authors reported having no relevant financial disclosures.

BOSTON – CT scans appear to be effective for detecting local recurrences and pulmonary metastases in patients treated for soft-tissue sarcomas of the extremities, for about a third less than the cost of follow-up with MRI.

In a retrospective study by Dr. Allison Maciver and her colleagues, among 91 patients with soft-tissue sarcomas of the extremity followed with CT, 11 patients had a total of 14 local recurrences detected on CT, and 11 of the recurrences were in patients who were clinically asymptomatic.

Surveillance CT also identified 15 cases of pulmonary metastases, and 4 incidental second primary malignancies, Dr. Maciver of the Roswell Park Cancer Institute in Buffalo, N.Y., and her coinvestigators found, and there was only one false-positive recurrence.

The benefits of CT over extremity MRI in this population include decreased imaging time, lower cost, and a larger field of view, allowing for detection of second primary malignancies, she noted in a poster session at the annual Society of Surgical Oncology Cancer Symposium.

Many sarcomas of the extremities are highly aggressive, and timely detection of local recurrences could improve chances for limb-sparing salvage therapies. Although MRI has typically been used to follow patients with sarcomas, it is expensive and has a limited field of view, Dr. Maciver said.

In addition, the risk of pulmonary metastases with some soft-tissue sarcomas is high, necessitating the use of chest CT as a surveillance tool.

To see whether CT scans of the chest and extremities could be a cost-effective surveillance strategy for both local recurrences and pulmonary metastases, the investigators did a retrospective study of a prospective database of patients who underwent surgical resection for soft-tissue sarcomas of the extremities from 2001 through 2014 and who had CT as the primary follow-up imaging modality.

They identified a total of 91 high-risk patients followed for a median of 50.5 months. The patients had an estimated 5-year freedom from local recurrence of 82%, and from distant recurrence of 80%. Five-year overall survival was 76%.

Of the 15 patients found on CT to have pulmonary metastases, there were 4 incidentally discovered second primary cancers, including 1 each of non–small cell lung cancer, pancreatic adenocarcinoma, Merkel cell carcinomatosis, and myxofibrosarcoma. There were no false-positive pulmonary metastases.

The estimated cost of 10 years of surveillance, based on 2014 gross technical costs, was $64,969 per patient for chest CT and extremity MRI, compared with $41,595 per patient for chest and extremity CT surveillance, a potential cost savings with the CT-only strategy of $23,374 per patient.

The investigators said that the overall benefits of CT, including the cost savings in an accountable care organization model, “appear to outweigh the slightly increased radiation exposure.”

The study was internally funded. The authors reported having no relevant financial disclosures.

References

References

Publications
Publications
Topics
Article Type
Display Headline
CT of chest, extremity effective for sarcoma follow-up
Display Headline
CT of chest, extremity effective for sarcoma follow-up
Click for Credit Status
Active
Sections
Article Source

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Lower-cost CT scans of the extremity and chest appear to be effective for surveillance of patients following resection of soft-tissue sarcomas.

Major finding: Of 91 patients with soft-tissue sarcomas of the extremity followed with CT, 11 had a total of 14 local recurrences detected. Of the recurrences, 11 were clinically asymptomatic.

Data source: A retrospective study of a prospectively maintained surgical database.

Disclosures: The study was internally funded. The authors reported having no relevant financial disclosures.

Lifetime HIV infection risk declines, but some still at high risk

Article Type
Changed
Fri, 01/18/2019 - 15:48
Display Headline
Lifetime HIV infection risk declines, but some still at high risk

BOSTON – The risk that a person living in the United States will become infected with HIV in his or her lifetime is declining compared with a decade ago, but the risk remains quite high among blacks and Hispanics/Latinos, investigators from the Centers for Disease Control and Prevention caution.

In fact, an estimated one in two black men who have sex with men (MSM) and one in four Hispanic/Latino MSM will be diagnosed with HIV within their lifetimes, reported CDC investigator Dr. Kristen Hess, at a media briefing and in an oral session at the 2016 Conference on Retroviruses and Opportunistic Infections.

Dr. Kristen Hess

“In the current analysis, we estimated that the overall lifetime risk is 1 in 99, which is reduced from 1 in 78 in the previous estimate [2004-2005]. However, there are vast disparities that still persist,” she said.

Estimates of lifetime risk are frequently used by epidemiologists to compare the burden of diseases such as cancer across populations, but are infrequently used for estimating lifetime risk of HIV infection, Dr. Hess said.

“The advantage to using lifetime risk is that it is more easily understood by the general population. Therefore, it could be a useful tool for clinicians, outreach workers, and policy makers,” she said in her podium presentation.

Previous estimates of lifetime HIV infection risk were based on data from 2004 and 2005, before data from all 50 states were available, leading the investigators to calculate new estimates based on more current information.

They drew on the National HIV Surveillance System for information about HIV diagnosis, mortality data from the National Center for Health Statistics, and U.S. Census data on population size by age, race/ethnicity, and state. The study covered the years 2009 through 2013.

The investigators used the number of HIV diagnoses and the number of non-HIV deaths to calculate the probability of a diagnosis of HIV at a given age and applied the probabilities to a hypothetical cohort to determine risk estimates.

They defined lifetime risk as” the cumulative probability of being diagnosed with HIV from birth.”

The lifetime risk of an HIV diagnosis for MSM varied widely by race/ethnicity. Overall, 1 in 64 men will be diagnosed with HIV during his lifetime, with the risk highest for black men at 1 in 20, followed by Hispanics at 1 in 48, native Hawaiian or other Pacific Islanders (NHOPI) at 1 in 82, American Indian/Alaskan Natives (AI/AN) at 1 in 129, whites at 1 in 132, and Asians with the lowest risk at 1 in 174.

For women the risks were considerably lower overall at 1 in 227. However, African American women had the same level of risk as Hispanic MSM (1 in 48). Risks for other racial/ethnic groups among women are: Hispanics, 1 in 227; NHOPI, 1 in 385; AI/AN, 1 in 399; whites, 1 in 880; and Asians, 1 in 883.

Lifetime risk from age 13 of an HIV diagnosis by risk group was highest among MSM, at 1 in 6, followed by female injectable drug users (1 in 23), male injectable drug users (1 in 36), heterosexual women (1 in 241), and heterosexual men (1 in 473).

Among MSM, the risk was highest for black men (1 in 2), followed by Hispanic/Latino (1 in 4), NHOPI (1 in 7), white (1 in 11), AI/AN (1 in 12) and Asians (1 in 14).

The overall lifetime risk of an HIV diagnosis among the U.S. population as a whole was 1 in 99. The area with the highest lifetime risk was Washington, D.C., with a risk of 1 in 13. Dr. Hess cautioned that the D.C. is a city, not a state, which could skew comparisons. The states with the highest lifetime HIV infection risk were Maryland, Georgia, and Florida, and in general, the highest lifetime risks were in the South.

The lowest overall risk was in North Dakota, at 1 in 670.

The investigators noted that their study was limited by its reliance on diagnosis rather than incidence data and may thus underestimate some risk categories because infection may occur several years before a diagnosis. Additionally, death certificates may be missing HIV information, she said.

“One of the goals of National HIV/AIDS strategy is to reduce disparities. Lifetime risk highlights work that still needs to be done and could be used to track progress toward this goal,” Dr. Hess said.

The study was supported by the CDC. Dr. Hess is a CDC employee.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
HIV, AIDS
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

BOSTON – The risk that a person living in the United States will become infected with HIV in his or her lifetime is declining compared with a decade ago, but the risk remains quite high among blacks and Hispanics/Latinos, investigators from the Centers for Disease Control and Prevention caution.

In fact, an estimated one in two black men who have sex with men (MSM) and one in four Hispanic/Latino MSM will be diagnosed with HIV within their lifetimes, reported CDC investigator Dr. Kristen Hess, at a media briefing and in an oral session at the 2016 Conference on Retroviruses and Opportunistic Infections.

Dr. Kristen Hess

“In the current analysis, we estimated that the overall lifetime risk is 1 in 99, which is reduced from 1 in 78 in the previous estimate [2004-2005]. However, there are vast disparities that still persist,” she said.

Estimates of lifetime risk are frequently used by epidemiologists to compare the burden of diseases such as cancer across populations, but are infrequently used for estimating lifetime risk of HIV infection, Dr. Hess said.

“The advantage to using lifetime risk is that it is more easily understood by the general population. Therefore, it could be a useful tool for clinicians, outreach workers, and policy makers,” she said in her podium presentation.

Previous estimates of lifetime HIV infection risk were based on data from 2004 and 2005, before data from all 50 states were available, leading the investigators to calculate new estimates based on more current information.

They drew on the National HIV Surveillance System for information about HIV diagnosis, mortality data from the National Center for Health Statistics, and U.S. Census data on population size by age, race/ethnicity, and state. The study covered the years 2009 through 2013.

The investigators used the number of HIV diagnoses and the number of non-HIV deaths to calculate the probability of a diagnosis of HIV at a given age and applied the probabilities to a hypothetical cohort to determine risk estimates.

They defined lifetime risk as” the cumulative probability of being diagnosed with HIV from birth.”

The lifetime risk of an HIV diagnosis for MSM varied widely by race/ethnicity. Overall, 1 in 64 men will be diagnosed with HIV during his lifetime, with the risk highest for black men at 1 in 20, followed by Hispanics at 1 in 48, native Hawaiian or other Pacific Islanders (NHOPI) at 1 in 82, American Indian/Alaskan Natives (AI/AN) at 1 in 129, whites at 1 in 132, and Asians with the lowest risk at 1 in 174.

For women the risks were considerably lower overall at 1 in 227. However, African American women had the same level of risk as Hispanic MSM (1 in 48). Risks for other racial/ethnic groups among women are: Hispanics, 1 in 227; NHOPI, 1 in 385; AI/AN, 1 in 399; whites, 1 in 880; and Asians, 1 in 883.

Lifetime risk from age 13 of an HIV diagnosis by risk group was highest among MSM, at 1 in 6, followed by female injectable drug users (1 in 23), male injectable drug users (1 in 36), heterosexual women (1 in 241), and heterosexual men (1 in 473).

Among MSM, the risk was highest for black men (1 in 2), followed by Hispanic/Latino (1 in 4), NHOPI (1 in 7), white (1 in 11), AI/AN (1 in 12) and Asians (1 in 14).

The overall lifetime risk of an HIV diagnosis among the U.S. population as a whole was 1 in 99. The area with the highest lifetime risk was Washington, D.C., with a risk of 1 in 13. Dr. Hess cautioned that the D.C. is a city, not a state, which could skew comparisons. The states with the highest lifetime HIV infection risk were Maryland, Georgia, and Florida, and in general, the highest lifetime risks were in the South.

The lowest overall risk was in North Dakota, at 1 in 670.

The investigators noted that their study was limited by its reliance on diagnosis rather than incidence data and may thus underestimate some risk categories because infection may occur several years before a diagnosis. Additionally, death certificates may be missing HIV information, she said.

“One of the goals of National HIV/AIDS strategy is to reduce disparities. Lifetime risk highlights work that still needs to be done and could be used to track progress toward this goal,” Dr. Hess said.

The study was supported by the CDC. Dr. Hess is a CDC employee.

BOSTON – The risk that a person living in the United States will become infected with HIV in his or her lifetime is declining compared with a decade ago, but the risk remains quite high among blacks and Hispanics/Latinos, investigators from the Centers for Disease Control and Prevention caution.

In fact, an estimated one in two black men who have sex with men (MSM) and one in four Hispanic/Latino MSM will be diagnosed with HIV within their lifetimes, reported CDC investigator Dr. Kristen Hess, at a media briefing and in an oral session at the 2016 Conference on Retroviruses and Opportunistic Infections.

Dr. Kristen Hess

“In the current analysis, we estimated that the overall lifetime risk is 1 in 99, which is reduced from 1 in 78 in the previous estimate [2004-2005]. However, there are vast disparities that still persist,” she said.

Estimates of lifetime risk are frequently used by epidemiologists to compare the burden of diseases such as cancer across populations, but are infrequently used for estimating lifetime risk of HIV infection, Dr. Hess said.

“The advantage to using lifetime risk is that it is more easily understood by the general population. Therefore, it could be a useful tool for clinicians, outreach workers, and policy makers,” she said in her podium presentation.

Previous estimates of lifetime HIV infection risk were based on data from 2004 and 2005, before data from all 50 states were available, leading the investigators to calculate new estimates based on more current information.

They drew on the National HIV Surveillance System for information about HIV diagnosis, mortality data from the National Center for Health Statistics, and U.S. Census data on population size by age, race/ethnicity, and state. The study covered the years 2009 through 2013.

The investigators used the number of HIV diagnoses and the number of non-HIV deaths to calculate the probability of a diagnosis of HIV at a given age and applied the probabilities to a hypothetical cohort to determine risk estimates.

They defined lifetime risk as” the cumulative probability of being diagnosed with HIV from birth.”

The lifetime risk of an HIV diagnosis for MSM varied widely by race/ethnicity. Overall, 1 in 64 men will be diagnosed with HIV during his lifetime, with the risk highest for black men at 1 in 20, followed by Hispanics at 1 in 48, native Hawaiian or other Pacific Islanders (NHOPI) at 1 in 82, American Indian/Alaskan Natives (AI/AN) at 1 in 129, whites at 1 in 132, and Asians with the lowest risk at 1 in 174.

For women the risks were considerably lower overall at 1 in 227. However, African American women had the same level of risk as Hispanic MSM (1 in 48). Risks for other racial/ethnic groups among women are: Hispanics, 1 in 227; NHOPI, 1 in 385; AI/AN, 1 in 399; whites, 1 in 880; and Asians, 1 in 883.

Lifetime risk from age 13 of an HIV diagnosis by risk group was highest among MSM, at 1 in 6, followed by female injectable drug users (1 in 23), male injectable drug users (1 in 36), heterosexual women (1 in 241), and heterosexual men (1 in 473).

Among MSM, the risk was highest for black men (1 in 2), followed by Hispanic/Latino (1 in 4), NHOPI (1 in 7), white (1 in 11), AI/AN (1 in 12) and Asians (1 in 14).

The overall lifetime risk of an HIV diagnosis among the U.S. population as a whole was 1 in 99. The area with the highest lifetime risk was Washington, D.C., with a risk of 1 in 13. Dr. Hess cautioned that the D.C. is a city, not a state, which could skew comparisons. The states with the highest lifetime HIV infection risk were Maryland, Georgia, and Florida, and in general, the highest lifetime risks were in the South.

The lowest overall risk was in North Dakota, at 1 in 670.

The investigators noted that their study was limited by its reliance on diagnosis rather than incidence data and may thus underestimate some risk categories because infection may occur several years before a diagnosis. Additionally, death certificates may be missing HIV information, she said.

“One of the goals of National HIV/AIDS strategy is to reduce disparities. Lifetime risk highlights work that still needs to be done and could be used to track progress toward this goal,” Dr. Hess said.

The study was supported by the CDC. Dr. Hess is a CDC employee.

References

References

Publications
Publications
Topics
Article Type
Display Headline
Lifetime HIV infection risk declines, but some still at high risk
Display Headline
Lifetime HIV infection risk declines, but some still at high risk
Legacy Keywords
HIV, AIDS
Legacy Keywords
HIV, AIDS
Sections
Article Source

AT CROI 2016

PURLs Copyright

Inside the Article

Vitals

Key clinical point: The risk that a person living in the United States will become infected with HIV in his or her lifetime is declining, compared with a decade ago.

Major finding: Black men who have sex with men have an estimated one in two lifetime risk of HIV infection.

Data source: National HIV Surveillance System (for diagnosis data), National Center for Health Statistics (mortality data), and U.S. Census (data on population size by age, race/ethnicity, and state).

Disclosures: The study was supported by the CDC. Dr. Hess is a CDC employee.