LayerRx Mapping ID
629
Slot System
Featured Buckets
Featured Buckets Admin
Medscape Lead Concept
3005205

Striking racial/ethnic differences seen in RCC features

Article Type
Changed
Fri, 01/04/2019 - 14:26

In a southwestern U.S. population having renal cell carcinoma (RCC), patient and disease characteristics differ by race/ethnicity in ways that may have implications for prevention, diagnosis, prognosis, and treatment, finds a single-center cohort study.

Investigators led by Ken Batai, PhD, of University of Arizona, Tucson, retrospectively reviewed the medical records of 294 patients with RCC as their first cancer who underwent partial or radical nephrectomy: 151 European Americans, 95 Hispanic Americans, 22 Native Americans, 9 African Americans, and 17 other race/ethnicity. About 12% overall had metastases at presentation.

On average, compared with European Americans, Hispanic Americans were about 5 years younger at diagnosis (55.8 vs. 60.5) and had higher odds of diagnosis before the age of 50 (odds ratio, 2.77), according to results published in Clinical Genitourinary Cancer.

Native Americans were even younger (49.7) and had dramatically elevated odds of diagnosis before that age (odds ratio, 6.23).

Relative to their European American counterparts, Hispanic Americans less commonly smoked (30.5% vs 48.6%) and African Americans more commonly had chronic kidney disease (37.5% vs. 5.8%). Both groups had higher prevalence of diabetes (45.6% and 54.5% vs. 21.7%). In addition, Native Americans had higher body mass index (35.2 vs. 30.7).

Clear cell histology was seen in 78.8% of European Americans, but in 92.6% of Hispanic Americans (odds ratio, 2.79) and 86.4% of Native Americans. African Americans more commonly had stage III or IV disease at diagnosis (77.8% vs. 35.3%; odds ratio, 6.51), but the racial/ethnic groups did not differ significantly on grade, tumor size, or presence of necrosis.

Among the Hispanic American patients undergoing radical nephrectomy, disease was more commonly of stage III or IV at diagnosis in those who were aged 65 or older (odds ratio, 10.48) and those who spoke Spanish as their primary language (odds ratios, 4.61).

The reasons for the observed racial/ethnic disparities remain unclear, according to Dr. Batai and his coinvestigators. Nonetheless, “it is necessary to better understand the clinical characteristics of these underserved Hispanic American and Native American populations with high kidney cancer burden,” they wrote.

“Our findings can direct future research toward elucidating the difference in tumor behavior among the different ethnic groups and health care issues causing poor outcomes,” they concluded. The findings also “bring ... awareness to practitioners treating patients from these racial/ethnic minority groups regarding the clinical characteristics and underlying issues in these patient populations.”

The study was supported by the American Cancer Society and the Partnership for Native American Cancer Prevention.

SOURCE: Batai K et al. Clin Genitourin Cancer. 2018 Oct 26. doi: 10.1016/j.clgc.2018.10.012.

Publications
Topics
Sections

In a southwestern U.S. population having renal cell carcinoma (RCC), patient and disease characteristics differ by race/ethnicity in ways that may have implications for prevention, diagnosis, prognosis, and treatment, finds a single-center cohort study.

Investigators led by Ken Batai, PhD, of University of Arizona, Tucson, retrospectively reviewed the medical records of 294 patients with RCC as their first cancer who underwent partial or radical nephrectomy: 151 European Americans, 95 Hispanic Americans, 22 Native Americans, 9 African Americans, and 17 other race/ethnicity. About 12% overall had metastases at presentation.

On average, compared with European Americans, Hispanic Americans were about 5 years younger at diagnosis (55.8 vs. 60.5) and had higher odds of diagnosis before the age of 50 (odds ratio, 2.77), according to results published in Clinical Genitourinary Cancer.

Native Americans were even younger (49.7) and had dramatically elevated odds of diagnosis before that age (odds ratio, 6.23).

Relative to their European American counterparts, Hispanic Americans less commonly smoked (30.5% vs 48.6%) and African Americans more commonly had chronic kidney disease (37.5% vs. 5.8%). Both groups had higher prevalence of diabetes (45.6% and 54.5% vs. 21.7%). In addition, Native Americans had higher body mass index (35.2 vs. 30.7).

Clear cell histology was seen in 78.8% of European Americans, but in 92.6% of Hispanic Americans (odds ratio, 2.79) and 86.4% of Native Americans. African Americans more commonly had stage III or IV disease at diagnosis (77.8% vs. 35.3%; odds ratio, 6.51), but the racial/ethnic groups did not differ significantly on grade, tumor size, or presence of necrosis.

Among the Hispanic American patients undergoing radical nephrectomy, disease was more commonly of stage III or IV at diagnosis in those who were aged 65 or older (odds ratio, 10.48) and those who spoke Spanish as their primary language (odds ratios, 4.61).

The reasons for the observed racial/ethnic disparities remain unclear, according to Dr. Batai and his coinvestigators. Nonetheless, “it is necessary to better understand the clinical characteristics of these underserved Hispanic American and Native American populations with high kidney cancer burden,” they wrote.

“Our findings can direct future research toward elucidating the difference in tumor behavior among the different ethnic groups and health care issues causing poor outcomes,” they concluded. The findings also “bring ... awareness to practitioners treating patients from these racial/ethnic minority groups regarding the clinical characteristics and underlying issues in these patient populations.”

The study was supported by the American Cancer Society and the Partnership for Native American Cancer Prevention.

SOURCE: Batai K et al. Clin Genitourin Cancer. 2018 Oct 26. doi: 10.1016/j.clgc.2018.10.012.

In a southwestern U.S. population having renal cell carcinoma (RCC), patient and disease characteristics differ by race/ethnicity in ways that may have implications for prevention, diagnosis, prognosis, and treatment, finds a single-center cohort study.

Investigators led by Ken Batai, PhD, of University of Arizona, Tucson, retrospectively reviewed the medical records of 294 patients with RCC as their first cancer who underwent partial or radical nephrectomy: 151 European Americans, 95 Hispanic Americans, 22 Native Americans, 9 African Americans, and 17 other race/ethnicity. About 12% overall had metastases at presentation.

On average, compared with European Americans, Hispanic Americans were about 5 years younger at diagnosis (55.8 vs. 60.5) and had higher odds of diagnosis before the age of 50 (odds ratio, 2.77), according to results published in Clinical Genitourinary Cancer.

Native Americans were even younger (49.7) and had dramatically elevated odds of diagnosis before that age (odds ratio, 6.23).

Relative to their European American counterparts, Hispanic Americans less commonly smoked (30.5% vs 48.6%) and African Americans more commonly had chronic kidney disease (37.5% vs. 5.8%). Both groups had higher prevalence of diabetes (45.6% and 54.5% vs. 21.7%). In addition, Native Americans had higher body mass index (35.2 vs. 30.7).

Clear cell histology was seen in 78.8% of European Americans, but in 92.6% of Hispanic Americans (odds ratio, 2.79) and 86.4% of Native Americans. African Americans more commonly had stage III or IV disease at diagnosis (77.8% vs. 35.3%; odds ratio, 6.51), but the racial/ethnic groups did not differ significantly on grade, tumor size, or presence of necrosis.

Among the Hispanic American patients undergoing radical nephrectomy, disease was more commonly of stage III or IV at diagnosis in those who were aged 65 or older (odds ratio, 10.48) and those who spoke Spanish as their primary language (odds ratios, 4.61).

The reasons for the observed racial/ethnic disparities remain unclear, according to Dr. Batai and his coinvestigators. Nonetheless, “it is necessary to better understand the clinical characteristics of these underserved Hispanic American and Native American populations with high kidney cancer burden,” they wrote.

“Our findings can direct future research toward elucidating the difference in tumor behavior among the different ethnic groups and health care issues causing poor outcomes,” they concluded. The findings also “bring ... awareness to practitioners treating patients from these racial/ethnic minority groups regarding the clinical characteristics and underlying issues in these patient populations.”

The study was supported by the American Cancer Society and the Partnership for Native American Cancer Prevention.

SOURCE: Batai K et al. Clin Genitourin Cancer. 2018 Oct 26. doi: 10.1016/j.clgc.2018.10.012.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM CLINICAL GENITOURINARY CANCER

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

Key clinical point: Patient and disease characteristics of RCC in a southwestern U.S. population differ by race/ethnicity.

Major finding: Compared with European Americans, Hispanic Americans and Native Americans were younger at diagnosis (55.8 and 49.7 vs. 60.5 years) and more often had clear cell histology (92.6% and 86.4% vs. 78.8%), and African Americans more often had stage III/IV disease (77.8% vs. 35.3%).

Study details: U.S. single-center retrospective cohort study of 294 patients with RCC who underwent partial or radical nephrectomy.

Disclosures: The authors declared that they did not have any conflicts of interest. The study was supported by the American Cancer Society and the Partnership for Native American Cancer Prevention.

Source: Batai K et al. Clin Genitourin Cancer. 2018 Oct 26. doi: 10.1016/j.clgc.2018.10.012.

Disqus Comments
Default
Use ProPublica

KPNA2 is “novel prognostic factor” in RCC

Article Type
Changed
Fri, 01/04/2019 - 14:25

 

High expression of karyopherin alpha 2 (KPNA2), a carrier protein that helps shuttle cancer-associated proteins from the nucleus to the cytoplasm, is an adverse prognostic factor in patients with clear cell or papillary renal cell carcinoma (RCC), according to a retrospective cohort study.

Senior author Glen Kristiansen, MD, director of the Institute of Pathology at the University Hospital Bonn (Germany), and his colleagues assessed tumor levels of KPNA2 protein by immunohistochemistry in 240 clinic patients with RCC (217 with clear cell histology, 23 with papillary histology). They also assessed tumor levels of KPNA2 mRNA in 771 patients with RCC (481 with clear cell histology, 290 with papillary cell histology) using publicly available gene expression data from the Cancer Genome Atlas (CGA).

Overall, 19% of the clinic patients’ tumors showed high expression of KPNA2 protein, according to results reported in Clinical Genitourinary Cancer. In addition, 26% of the CGA patients’ tumors showed high expression of KPNA2 mRNA.

Among patients with clear cell RCC, those with high tumor levels of KPNA2 protein survived roughly half as long as counterparts with low levels or none (74 months vs. 171 months); the difference was significant in univariate analysis (P = .012) but not in multivariate analysis that included well-known prognostic factors (HR, 1.491; P = .237). On the other hand, those with high tumor levels of KPNA2 mRNA had an elevated risk of death in both univariate analysis (HR, 2.31; P less than .001) and multivariate analysis (HR, 1.45; P = .035).

Among patients with papillary RCC, tumor levels of KPNA2 protein were not significantly associated with survival. However, those with high tumor levels of KPNA2 mRNA had a sharply elevated risk of death in both univariate analysis (HR, 9.7; P less than .001) and multivariate analysis (HR, 6.2; P = .004).

KPNA2 expression “represents a novel prognostic factor in these subtypes of RCC,” concluded Dr. Kristiansen and his coinvestigators. Therefore, this biomarker “could be used to stratify risk groups within RCC.” Collectively, the study’s findings suggest that KPNA2 is involved in both the pathogenesis and the progression of RCC. Given evidence that it helps transport p53 and fibroblast growth factor 2 at least in tumors with clear cell histology, “investigation of the nucleocytoplasmic transport through KPNA2 in [clear cell] RCC is an important task for future studies to better understand the link between elevated KPNA2 expression and altered biological processes like proliferation, cell growth, migration, invasion and tumor formation in RCC. In addition, examination of other members of [the] karyopherin-alpha family could elucidate the role of the nucleocytoplasmic transport system in pathogenesis of RCC.”

The authors reported that they had no conflict of interests.

SOURCE: Kristiansen G et al. Clin Genitourin Cancer. 2018 Oct 22. doi: 10.1016/j.clgc.2018.10.008.

Publications
Topics
Sections

 

High expression of karyopherin alpha 2 (KPNA2), a carrier protein that helps shuttle cancer-associated proteins from the nucleus to the cytoplasm, is an adverse prognostic factor in patients with clear cell or papillary renal cell carcinoma (RCC), according to a retrospective cohort study.

Senior author Glen Kristiansen, MD, director of the Institute of Pathology at the University Hospital Bonn (Germany), and his colleagues assessed tumor levels of KPNA2 protein by immunohistochemistry in 240 clinic patients with RCC (217 with clear cell histology, 23 with papillary histology). They also assessed tumor levels of KPNA2 mRNA in 771 patients with RCC (481 with clear cell histology, 290 with papillary cell histology) using publicly available gene expression data from the Cancer Genome Atlas (CGA).

Overall, 19% of the clinic patients’ tumors showed high expression of KPNA2 protein, according to results reported in Clinical Genitourinary Cancer. In addition, 26% of the CGA patients’ tumors showed high expression of KPNA2 mRNA.

Among patients with clear cell RCC, those with high tumor levels of KPNA2 protein survived roughly half as long as counterparts with low levels or none (74 months vs. 171 months); the difference was significant in univariate analysis (P = .012) but not in multivariate analysis that included well-known prognostic factors (HR, 1.491; P = .237). On the other hand, those with high tumor levels of KPNA2 mRNA had an elevated risk of death in both univariate analysis (HR, 2.31; P less than .001) and multivariate analysis (HR, 1.45; P = .035).

Among patients with papillary RCC, tumor levels of KPNA2 protein were not significantly associated with survival. However, those with high tumor levels of KPNA2 mRNA had a sharply elevated risk of death in both univariate analysis (HR, 9.7; P less than .001) and multivariate analysis (HR, 6.2; P = .004).

KPNA2 expression “represents a novel prognostic factor in these subtypes of RCC,” concluded Dr. Kristiansen and his coinvestigators. Therefore, this biomarker “could be used to stratify risk groups within RCC.” Collectively, the study’s findings suggest that KPNA2 is involved in both the pathogenesis and the progression of RCC. Given evidence that it helps transport p53 and fibroblast growth factor 2 at least in tumors with clear cell histology, “investigation of the nucleocytoplasmic transport through KPNA2 in [clear cell] RCC is an important task for future studies to better understand the link between elevated KPNA2 expression and altered biological processes like proliferation, cell growth, migration, invasion and tumor formation in RCC. In addition, examination of other members of [the] karyopherin-alpha family could elucidate the role of the nucleocytoplasmic transport system in pathogenesis of RCC.”

The authors reported that they had no conflict of interests.

SOURCE: Kristiansen G et al. Clin Genitourin Cancer. 2018 Oct 22. doi: 10.1016/j.clgc.2018.10.008.

 

High expression of karyopherin alpha 2 (KPNA2), a carrier protein that helps shuttle cancer-associated proteins from the nucleus to the cytoplasm, is an adverse prognostic factor in patients with clear cell or papillary renal cell carcinoma (RCC), according to a retrospective cohort study.

Senior author Glen Kristiansen, MD, director of the Institute of Pathology at the University Hospital Bonn (Germany), and his colleagues assessed tumor levels of KPNA2 protein by immunohistochemistry in 240 clinic patients with RCC (217 with clear cell histology, 23 with papillary histology). They also assessed tumor levels of KPNA2 mRNA in 771 patients with RCC (481 with clear cell histology, 290 with papillary cell histology) using publicly available gene expression data from the Cancer Genome Atlas (CGA).

Overall, 19% of the clinic patients’ tumors showed high expression of KPNA2 protein, according to results reported in Clinical Genitourinary Cancer. In addition, 26% of the CGA patients’ tumors showed high expression of KPNA2 mRNA.

Among patients with clear cell RCC, those with high tumor levels of KPNA2 protein survived roughly half as long as counterparts with low levels or none (74 months vs. 171 months); the difference was significant in univariate analysis (P = .012) but not in multivariate analysis that included well-known prognostic factors (HR, 1.491; P = .237). On the other hand, those with high tumor levels of KPNA2 mRNA had an elevated risk of death in both univariate analysis (HR, 2.31; P less than .001) and multivariate analysis (HR, 1.45; P = .035).

Among patients with papillary RCC, tumor levels of KPNA2 protein were not significantly associated with survival. However, those with high tumor levels of KPNA2 mRNA had a sharply elevated risk of death in both univariate analysis (HR, 9.7; P less than .001) and multivariate analysis (HR, 6.2; P = .004).

KPNA2 expression “represents a novel prognostic factor in these subtypes of RCC,” concluded Dr. Kristiansen and his coinvestigators. Therefore, this biomarker “could be used to stratify risk groups within RCC.” Collectively, the study’s findings suggest that KPNA2 is involved in both the pathogenesis and the progression of RCC. Given evidence that it helps transport p53 and fibroblast growth factor 2 at least in tumors with clear cell histology, “investigation of the nucleocytoplasmic transport through KPNA2 in [clear cell] RCC is an important task for future studies to better understand the link between elevated KPNA2 expression and altered biological processes like proliferation, cell growth, migration, invasion and tumor formation in RCC. In addition, examination of other members of [the] karyopherin-alpha family could elucidate the role of the nucleocytoplasmic transport system in pathogenesis of RCC.”

The authors reported that they had no conflict of interests.

SOURCE: Kristiansen G et al. Clin Genitourin Cancer. 2018 Oct 22. doi: 10.1016/j.clgc.2018.10.008.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM CLINICAL GENITOURINARY CANCER

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: High KPNA2 expression is associated with increased risk of death in clear cell and papillary renal cell carcinoma (RCC).

Major finding: Survival was poorer for patients with clear cell RCC having high KPNA2 protein levels (74 vs. 171 months) or mRNA levels (hazard ratio for death, 1.45) and for patients with papillary RCC having high KPNA2 mRNA levels (HR, 6.2).

Study details: A retrospective dual cohort study of 240 clinic patients and 771 Cancer Genome Atlas patients.

Disclosures: The authors reported that they had no conflict of interests.

Source: Kristiansen G et al. Clin Genitourin Cancer. 2018 Oct 22. doi: 10.1016/j.clgc.2018.10.008.

Disqus Comments
Default
Use ProPublica

ATLAS: High-risk RCC patients might benefit from adjuvant axitinib

Article Type
Changed
Fri, 01/04/2019 - 14:25

 

While adjuvant axitinib failed to improve disease-free survival in a recent phase 3 renal cell carcinoma (RCC) trial, the highest-risk subgroup appeared to benefit, according to a report on the study.

The phase 3 ATLAS trial was stopped early because of a lack of benefit for axitinib versus placebo in the study, which included patients with locoregional RCC at risk of recurrence after nephrectomy.

However, a prespecified analysis showed that axitinib reduced risk of disease-free survival events by about one-third in the highest-risk subset of patients, according to investigator David I. Quinn, MD, USC Norris Comprehensive Cancer Center, Los Angeles, and colleagues.

That finding tracks with results of the earlier S-TRAC trial, in which patients at high risk of tumor recurrence after nephrectomy had significantly longer disease-free survival with sunitinib versus placebo, Dr. Quinn and coauthors said.

“Taken together, these results support that patients at highest risk for RCC recurrence benefit from adjuvant treatment,” they wrote in Annals of Oncology.

In the ATLAS trial, Dr. Quinn and coinvestigators at 137 centers in eight countries enrolled 724 adults with newly diagnosed renal cell carcinoma (greater than or equal to pT2 and/or N+, any Fuhrman grade) with Eastern Cooperative Oncology Group status of 0 or 1 and prior nephrectomy.

Patients were randomly assigned to oral, twice-daily axitinib 5 mg or placebo for up to 3 years of treatment, and at least 1 year of treatment provided there was no recurrence, substantial toxicity, or withdrawal of consent.

For the primary endpoint, disease-free survival per independent review committee assessment, the hazard ratio was 0.870 (95% confidence interval, 0.660-1.147; P = .3211), according to the report. Disease-free survival as rated by investigators showed a somewhat larger but still not statistically significant reduction in risk of an event, Dr. Quinn and colleagues said.

However, in the prespecified subgroup analyses, the patients at highest risk (pT3 with Fuhrman grade greater than or equal to 3 or pT4 and/or N+, any T, any Fuhrman grade) had a reduction of risk with hazard ratios of 0.735 per independent review committee (P = .0704) and 0.641 per investigator (P = .0051).

The ATLAS study was designed before results of the S-TRAC study were known, and so patients at lower risk of recurrence were included, said Dr. Quinn and coauthors.

Ongoing trials are looking at sorafenib, everolimus, and immune checkpoint inhibitors in the adjuvant RCC setting, they noted in their discussion of ATLAS, S-TRAC, and other investigations.

Results from these trials may provide clarification on the future of adjuvant treatment for RCC, and whether angiogenesis inhibition is the key mechanism to obtain a reduction in risk of relapse after nephrectomy,” they said.

The study was sponsored by Pfizer and SFJ Pharmaceuticals. Dr. Quinn reported providing advisory board services for Pfizer, Bayer, Novartis, Bristol-Myers Squibb, Merck, Exelixis, Genentech, Roche, AstraZeneca, and Astellas. Coauthors reported disclosures related to Bristol-Myers Squibb, Ipsen, MSD, Novartis, Pfizer, and Roche, among others.

SOURCE: Quinn DI et al. Ann Oncol. 2018 Oct 20. doi: 10.1093/annonc/mdy454.

Publications
Topics
Sections

 

While adjuvant axitinib failed to improve disease-free survival in a recent phase 3 renal cell carcinoma (RCC) trial, the highest-risk subgroup appeared to benefit, according to a report on the study.

The phase 3 ATLAS trial was stopped early because of a lack of benefit for axitinib versus placebo in the study, which included patients with locoregional RCC at risk of recurrence after nephrectomy.

However, a prespecified analysis showed that axitinib reduced risk of disease-free survival events by about one-third in the highest-risk subset of patients, according to investigator David I. Quinn, MD, USC Norris Comprehensive Cancer Center, Los Angeles, and colleagues.

That finding tracks with results of the earlier S-TRAC trial, in which patients at high risk of tumor recurrence after nephrectomy had significantly longer disease-free survival with sunitinib versus placebo, Dr. Quinn and coauthors said.

“Taken together, these results support that patients at highest risk for RCC recurrence benefit from adjuvant treatment,” they wrote in Annals of Oncology.

In the ATLAS trial, Dr. Quinn and coinvestigators at 137 centers in eight countries enrolled 724 adults with newly diagnosed renal cell carcinoma (greater than or equal to pT2 and/or N+, any Fuhrman grade) with Eastern Cooperative Oncology Group status of 0 or 1 and prior nephrectomy.

Patients were randomly assigned to oral, twice-daily axitinib 5 mg or placebo for up to 3 years of treatment, and at least 1 year of treatment provided there was no recurrence, substantial toxicity, or withdrawal of consent.

For the primary endpoint, disease-free survival per independent review committee assessment, the hazard ratio was 0.870 (95% confidence interval, 0.660-1.147; P = .3211), according to the report. Disease-free survival as rated by investigators showed a somewhat larger but still not statistically significant reduction in risk of an event, Dr. Quinn and colleagues said.

However, in the prespecified subgroup analyses, the patients at highest risk (pT3 with Fuhrman grade greater than or equal to 3 or pT4 and/or N+, any T, any Fuhrman grade) had a reduction of risk with hazard ratios of 0.735 per independent review committee (P = .0704) and 0.641 per investigator (P = .0051).

The ATLAS study was designed before results of the S-TRAC study were known, and so patients at lower risk of recurrence were included, said Dr. Quinn and coauthors.

Ongoing trials are looking at sorafenib, everolimus, and immune checkpoint inhibitors in the adjuvant RCC setting, they noted in their discussion of ATLAS, S-TRAC, and other investigations.

Results from these trials may provide clarification on the future of adjuvant treatment for RCC, and whether angiogenesis inhibition is the key mechanism to obtain a reduction in risk of relapse after nephrectomy,” they said.

The study was sponsored by Pfizer and SFJ Pharmaceuticals. Dr. Quinn reported providing advisory board services for Pfizer, Bayer, Novartis, Bristol-Myers Squibb, Merck, Exelixis, Genentech, Roche, AstraZeneca, and Astellas. Coauthors reported disclosures related to Bristol-Myers Squibb, Ipsen, MSD, Novartis, Pfizer, and Roche, among others.

SOURCE: Quinn DI et al. Ann Oncol. 2018 Oct 20. doi: 10.1093/annonc/mdy454.

 

While adjuvant axitinib failed to improve disease-free survival in a recent phase 3 renal cell carcinoma (RCC) trial, the highest-risk subgroup appeared to benefit, according to a report on the study.

The phase 3 ATLAS trial was stopped early because of a lack of benefit for axitinib versus placebo in the study, which included patients with locoregional RCC at risk of recurrence after nephrectomy.

However, a prespecified analysis showed that axitinib reduced risk of disease-free survival events by about one-third in the highest-risk subset of patients, according to investigator David I. Quinn, MD, USC Norris Comprehensive Cancer Center, Los Angeles, and colleagues.

That finding tracks with results of the earlier S-TRAC trial, in which patients at high risk of tumor recurrence after nephrectomy had significantly longer disease-free survival with sunitinib versus placebo, Dr. Quinn and coauthors said.

“Taken together, these results support that patients at highest risk for RCC recurrence benefit from adjuvant treatment,” they wrote in Annals of Oncology.

In the ATLAS trial, Dr. Quinn and coinvestigators at 137 centers in eight countries enrolled 724 adults with newly diagnosed renal cell carcinoma (greater than or equal to pT2 and/or N+, any Fuhrman grade) with Eastern Cooperative Oncology Group status of 0 or 1 and prior nephrectomy.

Patients were randomly assigned to oral, twice-daily axitinib 5 mg or placebo for up to 3 years of treatment, and at least 1 year of treatment provided there was no recurrence, substantial toxicity, or withdrawal of consent.

For the primary endpoint, disease-free survival per independent review committee assessment, the hazard ratio was 0.870 (95% confidence interval, 0.660-1.147; P = .3211), according to the report. Disease-free survival as rated by investigators showed a somewhat larger but still not statistically significant reduction in risk of an event, Dr. Quinn and colleagues said.

However, in the prespecified subgroup analyses, the patients at highest risk (pT3 with Fuhrman grade greater than or equal to 3 or pT4 and/or N+, any T, any Fuhrman grade) had a reduction of risk with hazard ratios of 0.735 per independent review committee (P = .0704) and 0.641 per investigator (P = .0051).

The ATLAS study was designed before results of the S-TRAC study were known, and so patients at lower risk of recurrence were included, said Dr. Quinn and coauthors.

Ongoing trials are looking at sorafenib, everolimus, and immune checkpoint inhibitors in the adjuvant RCC setting, they noted in their discussion of ATLAS, S-TRAC, and other investigations.

Results from these trials may provide clarification on the future of adjuvant treatment for RCC, and whether angiogenesis inhibition is the key mechanism to obtain a reduction in risk of relapse after nephrectomy,” they said.

The study was sponsored by Pfizer and SFJ Pharmaceuticals. Dr. Quinn reported providing advisory board services for Pfizer, Bayer, Novartis, Bristol-Myers Squibb, Merck, Exelixis, Genentech, Roche, AstraZeneca, and Astellas. Coauthors reported disclosures related to Bristol-Myers Squibb, Ipsen, MSD, Novartis, Pfizer, and Roche, among others.

SOURCE: Quinn DI et al. Ann Oncol. 2018 Oct 20. doi: 10.1093/annonc/mdy454.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM ANNALS OF ONCOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: While adjuvant axitinib failed to improve disease-free survival in a recent phase 3 renal cell carcinoma trial, the highest-risk subgroup appeared to benefit.

Major finding: The highest-risk subpopulation (pT3 with Fuhrman grade greater than or equal to 3 or pT4 and/or N+, any T, any Fuhrman grade) had a reduction of risk per assessments by independent review committee (HR, 0.735; P = .0704) and investigators (HR, 0.641; P = .0051).

Study details: Results from ATLAS, a phase 3, randomized trial including 724 patients with locoregional RCC.

Disclosures: The study was sponsored by Pfizer and SFJ Pharmaceuticals. Study authors reported disclosures related to Pfizer, Bayer, Novartis, Bristol-Myers Squibb, Merck, Exelixis, Genentech, Roche, AstraZeneca, and Astellas, among others.

Source: Quinn DI et al. Ann Oncol. 2018 Oct 20. doi: 10.1093/annonc/mdy454.

Disqus Comments
Default
Use ProPublica

Checkpoint inhibitor/TKI combo improves PFS of RCC over sunitinib

Article Type
Changed
Fri, 01/04/2019 - 14:25

MUNICH – The combination of the immune checkpoint inhibitor avelumab (Bavencio) and the tyrosine kinase inhibitor axitinib (Inlyta) was associated with longer progression-free survival and higher objective response rates than was sunitinib as frontline therapy for patients with advanced renal cell carcinoma, investigators found.

Neil Osterweil/MDedge News
Dr. Robert J. Motzer

The progression-free survival (PFS) benefit of the combination was seen both in the subset of patients with tumors expressing programmed death-1 ligand 1 (PD-L1) on at least 1% of their cells as well as in the overall study population, reported Robert J. Motzer, MD, of Memorial Sloan Kettering Cancer Center, New York, on behalf of colleagues in the JAVELIN Renal 101 trial.

“This was very early on, at an interim analysis, showing a profound effect,” he said at a briefing prior to his presentation of the data in a presidential symposium at the European Society for Medical Oncology Congress.

“The results support this being a new first-line standard of care, and possibly a first-line option for advanced RCC based on these data,” he added.

Briefing discussant John Haanen, PhD, of the Netherlands Cancer Institute in Amsterdam, said that “based on preclinical data, it makes sense to combine angiogenesis inhibitors and immunotherapy, because we know angiogenesis, VEGF [vascular endothelial growth factor] especially impacts on the way the immune system can respond to the kidney cancer, and by taking away some of these negative effects of the VEGF by using an anti-VEGF drug, the immunotherapy may work better,” he said.

Neil Osterweil/MDedge News
Dr. John Haanen


“This is the first study of a TKI plus and anti-PD-L1 drug showing an improved progression-free survival in this patient population,” he added.

On Oct. 18, the day before the start of ESMO 2018, Merck announced in a press release positive results of a phase 3 trial comparing axitinib in combination with a different checkpoint inhibitor, pembrolizumab (Keytruda), compared with sunitinib monotherapy. The press release, typically sparse on details, said that the trial met both primary endpoints of overall survival (OS) and PFS in the first-line treatment of advanced or metastatic RCC.

In the JAVELIN Renal 101 trial, investigators enrolled and randomized 886 patients, 873 of whom went on to treatment: 434 assigned to avelumab plus axitinib, and 439 to sunitinib.

A total of 560 patients were determined to be PD-L1-positive according to the Ventana assay: 270 assigned to the combination, and 290 assigned to sunitinib.

Avelumab was delivered 10 mg/kg intravenously every 2 weeks, and oral axitinib was given 5 mg twice daily over a 6-week cycle. Oral sunitinib was give 50 mg daily for 4 weeks, followed by 2 weeks off, for every cycle.

Median PFS by independent review in the PD-L1-positive patients, one of two primary endpoints, was 13.8 months in the avelumab/axitinib arm, vs. 7.2 months in the sunitinib arm. This translated into a stratified hazard ratio (HR) of 0.61 (P less than .001) favoring the combination.

The respective median PFS in the overall population was 13.8 vs. 8.4 months, respectively (HR, 0.69; P = .0001).

Objective response rates also were higher with the combination in both the PD-L1-positive population (55% vs. 26%), and in the overall population (51% vs. 26%). At the time of data cutoff, the median duration of response had not been reached in either treatment arm in either population.

The stratified odds ratio for response with avelumab/axitinib was 3.098 (P less than .001).

At the time of the data cutoff for this interim analysis, overall survival data were not mature. OS in the PD-L1-positive population, the second primary endpoint, will be reported at a later date, Dr. Motzer said.

The incidence of treatment-related adverse events was similar in the groups, at 95% of patients in the avelumab arm and 96% in the sunitinib arm. Grade 3 or 4 events occurred in 51% and 48% of patients respectively. Grade 3/4 hypertension was the highest-frequency event, occurring in 24% of patients on avelumab/axitinib vs. 15% on sunitinib.

Immune-related adverse events occurred in 38% of patients in the avelumab group, including hypothyroidism, liver function test abnormalities, adrenal insufficiency, acute kidney injury, colitis, and hepatotoxicity. Most of the events occurred in 1% or 2% of patients, except hypothyroidism, which occurred in 21%.

High-dose corticosteroids were administered to 11% of patients who experienced an immune-related adverse event.

“One of the beauties of this combination is really its tolerability,” said Viktor Grünwald, MD, of the West German Cancer Center in Essen, the invited discussant at the symposium.

“When it comes to toxicities, they are pretty much in the same range, which is really surprising,” he said.

He noted, however, that it’s still an open question whether the combination of avelumab and axitinib is better than sequencing of other agents, given the current absence of evidence of an OS or quality-of-life benefit for the combination.

At present, the best evidence supports the use of ipilimumab (Yervoy) and nivolumab (Opdivo), which provides a clinically relevant OS benefit in patients with intermediate and high-risk disease, and this therapy should remain the standard of care, Dr. Grünwald said.

“I do believe that there is a niche, in favorable-risk patients, for this specific combination [avelumab/axitinib],” he said.

The trial was sponsored by Pfizer as part of an alliance between Pfizer and Merck KGaA. Dr. Motzer disclosed consulting or advisory roles with Pfizer, Merck, and others, and research funding from Pfizer and others. Dr. Haanen disclosed financial compensation for advisory work he did for Pfizer, Merck, Sharpe & Dohme, and others, and grant support from Bristol-Myers Squibb, MSD, and Novartis. Dr. Grünwald disclosed honoraria for speaking and advising from MSD, and funding from Pfizer, MSD, and others.

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

MUNICH – The combination of the immune checkpoint inhibitor avelumab (Bavencio) and the tyrosine kinase inhibitor axitinib (Inlyta) was associated with longer progression-free survival and higher objective response rates than was sunitinib as frontline therapy for patients with advanced renal cell carcinoma, investigators found.

Neil Osterweil/MDedge News
Dr. Robert J. Motzer

The progression-free survival (PFS) benefit of the combination was seen both in the subset of patients with tumors expressing programmed death-1 ligand 1 (PD-L1) on at least 1% of their cells as well as in the overall study population, reported Robert J. Motzer, MD, of Memorial Sloan Kettering Cancer Center, New York, on behalf of colleagues in the JAVELIN Renal 101 trial.

“This was very early on, at an interim analysis, showing a profound effect,” he said at a briefing prior to his presentation of the data in a presidential symposium at the European Society for Medical Oncology Congress.

“The results support this being a new first-line standard of care, and possibly a first-line option for advanced RCC based on these data,” he added.

Briefing discussant John Haanen, PhD, of the Netherlands Cancer Institute in Amsterdam, said that “based on preclinical data, it makes sense to combine angiogenesis inhibitors and immunotherapy, because we know angiogenesis, VEGF [vascular endothelial growth factor] especially impacts on the way the immune system can respond to the kidney cancer, and by taking away some of these negative effects of the VEGF by using an anti-VEGF drug, the immunotherapy may work better,” he said.

Neil Osterweil/MDedge News
Dr. John Haanen


“This is the first study of a TKI plus and anti-PD-L1 drug showing an improved progression-free survival in this patient population,” he added.

On Oct. 18, the day before the start of ESMO 2018, Merck announced in a press release positive results of a phase 3 trial comparing axitinib in combination with a different checkpoint inhibitor, pembrolizumab (Keytruda), compared with sunitinib monotherapy. The press release, typically sparse on details, said that the trial met both primary endpoints of overall survival (OS) and PFS in the first-line treatment of advanced or metastatic RCC.

In the JAVELIN Renal 101 trial, investigators enrolled and randomized 886 patients, 873 of whom went on to treatment: 434 assigned to avelumab plus axitinib, and 439 to sunitinib.

A total of 560 patients were determined to be PD-L1-positive according to the Ventana assay: 270 assigned to the combination, and 290 assigned to sunitinib.

Avelumab was delivered 10 mg/kg intravenously every 2 weeks, and oral axitinib was given 5 mg twice daily over a 6-week cycle. Oral sunitinib was give 50 mg daily for 4 weeks, followed by 2 weeks off, for every cycle.

Median PFS by independent review in the PD-L1-positive patients, one of two primary endpoints, was 13.8 months in the avelumab/axitinib arm, vs. 7.2 months in the sunitinib arm. This translated into a stratified hazard ratio (HR) of 0.61 (P less than .001) favoring the combination.

The respective median PFS in the overall population was 13.8 vs. 8.4 months, respectively (HR, 0.69; P = .0001).

Objective response rates also were higher with the combination in both the PD-L1-positive population (55% vs. 26%), and in the overall population (51% vs. 26%). At the time of data cutoff, the median duration of response had not been reached in either treatment arm in either population.

The stratified odds ratio for response with avelumab/axitinib was 3.098 (P less than .001).

At the time of the data cutoff for this interim analysis, overall survival data were not mature. OS in the PD-L1-positive population, the second primary endpoint, will be reported at a later date, Dr. Motzer said.

The incidence of treatment-related adverse events was similar in the groups, at 95% of patients in the avelumab arm and 96% in the sunitinib arm. Grade 3 or 4 events occurred in 51% and 48% of patients respectively. Grade 3/4 hypertension was the highest-frequency event, occurring in 24% of patients on avelumab/axitinib vs. 15% on sunitinib.

Immune-related adverse events occurred in 38% of patients in the avelumab group, including hypothyroidism, liver function test abnormalities, adrenal insufficiency, acute kidney injury, colitis, and hepatotoxicity. Most of the events occurred in 1% or 2% of patients, except hypothyroidism, which occurred in 21%.

High-dose corticosteroids were administered to 11% of patients who experienced an immune-related adverse event.

“One of the beauties of this combination is really its tolerability,” said Viktor Grünwald, MD, of the West German Cancer Center in Essen, the invited discussant at the symposium.

“When it comes to toxicities, they are pretty much in the same range, which is really surprising,” he said.

He noted, however, that it’s still an open question whether the combination of avelumab and axitinib is better than sequencing of other agents, given the current absence of evidence of an OS or quality-of-life benefit for the combination.

At present, the best evidence supports the use of ipilimumab (Yervoy) and nivolumab (Opdivo), which provides a clinically relevant OS benefit in patients with intermediate and high-risk disease, and this therapy should remain the standard of care, Dr. Grünwald said.

“I do believe that there is a niche, in favorable-risk patients, for this specific combination [avelumab/axitinib],” he said.

The trial was sponsored by Pfizer as part of an alliance between Pfizer and Merck KGaA. Dr. Motzer disclosed consulting or advisory roles with Pfizer, Merck, and others, and research funding from Pfizer and others. Dr. Haanen disclosed financial compensation for advisory work he did for Pfizer, Merck, Sharpe & Dohme, and others, and grant support from Bristol-Myers Squibb, MSD, and Novartis. Dr. Grünwald disclosed honoraria for speaking and advising from MSD, and funding from Pfizer, MSD, and others.

MUNICH – The combination of the immune checkpoint inhibitor avelumab (Bavencio) and the tyrosine kinase inhibitor axitinib (Inlyta) was associated with longer progression-free survival and higher objective response rates than was sunitinib as frontline therapy for patients with advanced renal cell carcinoma, investigators found.

Neil Osterweil/MDedge News
Dr. Robert J. Motzer

The progression-free survival (PFS) benefit of the combination was seen both in the subset of patients with tumors expressing programmed death-1 ligand 1 (PD-L1) on at least 1% of their cells as well as in the overall study population, reported Robert J. Motzer, MD, of Memorial Sloan Kettering Cancer Center, New York, on behalf of colleagues in the JAVELIN Renal 101 trial.

“This was very early on, at an interim analysis, showing a profound effect,” he said at a briefing prior to his presentation of the data in a presidential symposium at the European Society for Medical Oncology Congress.

“The results support this being a new first-line standard of care, and possibly a first-line option for advanced RCC based on these data,” he added.

Briefing discussant John Haanen, PhD, of the Netherlands Cancer Institute in Amsterdam, said that “based on preclinical data, it makes sense to combine angiogenesis inhibitors and immunotherapy, because we know angiogenesis, VEGF [vascular endothelial growth factor] especially impacts on the way the immune system can respond to the kidney cancer, and by taking away some of these negative effects of the VEGF by using an anti-VEGF drug, the immunotherapy may work better,” he said.

Neil Osterweil/MDedge News
Dr. John Haanen


“This is the first study of a TKI plus and anti-PD-L1 drug showing an improved progression-free survival in this patient population,” he added.

On Oct. 18, the day before the start of ESMO 2018, Merck announced in a press release positive results of a phase 3 trial comparing axitinib in combination with a different checkpoint inhibitor, pembrolizumab (Keytruda), compared with sunitinib monotherapy. The press release, typically sparse on details, said that the trial met both primary endpoints of overall survival (OS) and PFS in the first-line treatment of advanced or metastatic RCC.

In the JAVELIN Renal 101 trial, investigators enrolled and randomized 886 patients, 873 of whom went on to treatment: 434 assigned to avelumab plus axitinib, and 439 to sunitinib.

A total of 560 patients were determined to be PD-L1-positive according to the Ventana assay: 270 assigned to the combination, and 290 assigned to sunitinib.

Avelumab was delivered 10 mg/kg intravenously every 2 weeks, and oral axitinib was given 5 mg twice daily over a 6-week cycle. Oral sunitinib was give 50 mg daily for 4 weeks, followed by 2 weeks off, for every cycle.

Median PFS by independent review in the PD-L1-positive patients, one of two primary endpoints, was 13.8 months in the avelumab/axitinib arm, vs. 7.2 months in the sunitinib arm. This translated into a stratified hazard ratio (HR) of 0.61 (P less than .001) favoring the combination.

The respective median PFS in the overall population was 13.8 vs. 8.4 months, respectively (HR, 0.69; P = .0001).

Objective response rates also were higher with the combination in both the PD-L1-positive population (55% vs. 26%), and in the overall population (51% vs. 26%). At the time of data cutoff, the median duration of response had not been reached in either treatment arm in either population.

The stratified odds ratio for response with avelumab/axitinib was 3.098 (P less than .001).

At the time of the data cutoff for this interim analysis, overall survival data were not mature. OS in the PD-L1-positive population, the second primary endpoint, will be reported at a later date, Dr. Motzer said.

The incidence of treatment-related adverse events was similar in the groups, at 95% of patients in the avelumab arm and 96% in the sunitinib arm. Grade 3 or 4 events occurred in 51% and 48% of patients respectively. Grade 3/4 hypertension was the highest-frequency event, occurring in 24% of patients on avelumab/axitinib vs. 15% on sunitinib.

Immune-related adverse events occurred in 38% of patients in the avelumab group, including hypothyroidism, liver function test abnormalities, adrenal insufficiency, acute kidney injury, colitis, and hepatotoxicity. Most of the events occurred in 1% or 2% of patients, except hypothyroidism, which occurred in 21%.

High-dose corticosteroids were administered to 11% of patients who experienced an immune-related adverse event.

“One of the beauties of this combination is really its tolerability,” said Viktor Grünwald, MD, of the West German Cancer Center in Essen, the invited discussant at the symposium.

“When it comes to toxicities, they are pretty much in the same range, which is really surprising,” he said.

He noted, however, that it’s still an open question whether the combination of avelumab and axitinib is better than sequencing of other agents, given the current absence of evidence of an OS or quality-of-life benefit for the combination.

At present, the best evidence supports the use of ipilimumab (Yervoy) and nivolumab (Opdivo), which provides a clinically relevant OS benefit in patients with intermediate and high-risk disease, and this therapy should remain the standard of care, Dr. Grünwald said.

“I do believe that there is a niche, in favorable-risk patients, for this specific combination [avelumab/axitinib],” he said.

The trial was sponsored by Pfizer as part of an alliance between Pfizer and Merck KGaA. Dr. Motzer disclosed consulting or advisory roles with Pfizer, Merck, and others, and research funding from Pfizer and others. Dr. Haanen disclosed financial compensation for advisory work he did for Pfizer, Merck, Sharpe & Dohme, and others, and grant support from Bristol-Myers Squibb, MSD, and Novartis. Dr. Grünwald disclosed honoraria for speaking and advising from MSD, and funding from Pfizer, MSD, and others.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ESMO 2018

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

Key clinical point: Progression-free survival was superior with avelumab and axitinib vs. sunitinib regardless of PD-L1-status.

Major finding: Median PFS by independent review in PD-L1-positive patients was 13.8 months with avelumab/axitinib vs. 7.2 months with sunitinib.

Study details: Prospective randomized phase 3 trial of 560 patients with advanced RCC.

Disclosures: The trial was sponsored by Pfizer as part of an alliance between Pfizer and Merck KGaA. Dr. Motzer disclosed consulting or advisory roles with Pfizer, Merck, and others, and research funding from Pfizer and others. Dr. Haanen disclosed financial compensation for advisory work he did for Pfizer, Merck, Sharpe & Dohme, and others, and grant support from Bristol-Myers Squibb, MSD, and Novartis. Dr. Grünwald disclosed honoraria for speaking and advising from MSD, and funding from Pfizer, MSD, and others.

Source: Motzer RJ et al. ESMO 2018. Abstract LBA6_PR.

Disqus Comments
Default
Use ProPublica

mRCC: Pazopanib appears safe in renal failure

Article Type
Changed
Fri, 01/04/2019 - 14:25

There was no difference in the incidence of adverse events in a group of patients being treated with pazopanib for metastatic renal cell carcinoma with comorbid renal dysfunction.

In a multicenter, retrospective study that included 229 patients with or without renal insufficiency being treated for metastatic renal cell carcinoma with pazopanib, no significant differences were found in the incidence of adverse events between groups. Similar results were demonstrated for both efficacy parameters, progression-free survival, and overall survival (P = .6), Cristina Masini, MD, of AUSL-IRCCS in Reggio Emilia, Italy, and colleagues reported in Clinical Genitourinary Cancer.

The researchers also determined that dose reductions occurred more often in patients with renal insufficiency, compared with those that were renal competent (52% vs. 36%; P = .04).

The majority of study participants received a starting dose of 800 mg daily of pazopanib, which was reduced to a minimum of 200 mg daily in 19% of participants from the renal impairment group, compared with less than 1% in the nonrenally impaired group.

“The similar efficacy and safety displayed by pazopanib in patients with poor renal function, compared with those with normal function may have a major relevance for therapy individualization in clinical practice,” the investigators concluded, adding that, because of the retrospective study design, further research is needed to fully establish any causal links between pazopanib and renal insufficiency.

The authors reported that editorial assistance was supported by Novartis. No other conflict of interests were reported
 

SOURCE: Masini C et al. Clin Genitourin Cancer. 2018 Oct 1. doi: 10.1016/j.clgc.2018.10.001.

Publications
Topics
Sections

There was no difference in the incidence of adverse events in a group of patients being treated with pazopanib for metastatic renal cell carcinoma with comorbid renal dysfunction.

In a multicenter, retrospective study that included 229 patients with or without renal insufficiency being treated for metastatic renal cell carcinoma with pazopanib, no significant differences were found in the incidence of adverse events between groups. Similar results were demonstrated for both efficacy parameters, progression-free survival, and overall survival (P = .6), Cristina Masini, MD, of AUSL-IRCCS in Reggio Emilia, Italy, and colleagues reported in Clinical Genitourinary Cancer.

The researchers also determined that dose reductions occurred more often in patients with renal insufficiency, compared with those that were renal competent (52% vs. 36%; P = .04).

The majority of study participants received a starting dose of 800 mg daily of pazopanib, which was reduced to a minimum of 200 mg daily in 19% of participants from the renal impairment group, compared with less than 1% in the nonrenally impaired group.

“The similar efficacy and safety displayed by pazopanib in patients with poor renal function, compared with those with normal function may have a major relevance for therapy individualization in clinical practice,” the investigators concluded, adding that, because of the retrospective study design, further research is needed to fully establish any causal links between pazopanib and renal insufficiency.

The authors reported that editorial assistance was supported by Novartis. No other conflict of interests were reported
 

SOURCE: Masini C et al. Clin Genitourin Cancer. 2018 Oct 1. doi: 10.1016/j.clgc.2018.10.001.

There was no difference in the incidence of adverse events in a group of patients being treated with pazopanib for metastatic renal cell carcinoma with comorbid renal dysfunction.

In a multicenter, retrospective study that included 229 patients with or without renal insufficiency being treated for metastatic renal cell carcinoma with pazopanib, no significant differences were found in the incidence of adverse events between groups. Similar results were demonstrated for both efficacy parameters, progression-free survival, and overall survival (P = .6), Cristina Masini, MD, of AUSL-IRCCS in Reggio Emilia, Italy, and colleagues reported in Clinical Genitourinary Cancer.

The researchers also determined that dose reductions occurred more often in patients with renal insufficiency, compared with those that were renal competent (52% vs. 36%; P = .04).

The majority of study participants received a starting dose of 800 mg daily of pazopanib, which was reduced to a minimum of 200 mg daily in 19% of participants from the renal impairment group, compared with less than 1% in the nonrenally impaired group.

“The similar efficacy and safety displayed by pazopanib in patients with poor renal function, compared with those with normal function may have a major relevance for therapy individualization in clinical practice,” the investigators concluded, adding that, because of the retrospective study design, further research is needed to fully establish any causal links between pazopanib and renal insufficiency.

The authors reported that editorial assistance was supported by Novartis. No other conflict of interests were reported
 

SOURCE: Masini C et al. Clin Genitourin Cancer. 2018 Oct 1. doi: 10.1016/j.clgc.2018.10.001.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM CLINICAL GENITOURINARY CANCER

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

Key clinical point: Pazopanib may be safe and effective for metastatic renal cell carcinoma in patients with renal failure.

Major finding: No difference was reported in the incidence of adverse events in patients with or without renal dysfunction.

Study details: A retrospective analysis of 229 metastatic renal cell carcinoma patients treated with pazopanib.

Disclosures: Editorial assistance was supported by Novartis. The authors reported no conflict of interests related to the work.

Source: Masini C et al. Clin Genitourin Cancer. 2018 Oct 1. doi: 10.1016/j.clgc.2018.10.001.

Disqus Comments
Default
Use ProPublica

No ADT-dementia link in large VA prostate cancer cohort study

Article Type
Changed
Thu, 12/15/2022 - 14:44

In contrast to other recent studies, androgen deprivation therapy (ADT) had no link to dementia in a observational cohort study of more than 45,000 men with prostate cancer who received definitive radiotherapy, investigators have reported.

No significant associations were found between ADT and Alzheimer’s disease or vascular dementia, or between shorter or longer courses of ADT and any dementia studied, according to Rishi Deka, PhD, of Veterans Affairs San Diego Health Care System, La Jolla, Calif., and coinvestigators.

“These results may mitigate concerns regarding the long-term risks of ADT on cognitive health in the treatment of prostate cancer,” Dr. Deka and colleagues wrote in JAMA Oncology.

Two other recent studies showed strong, statistically significant associations between ADT and dementia in prostate cancer. However, those studies combined patients with local and metastatic disease, receiving ADT in the upfront or recurrent settings, while the present study looked specifically at men with nonmetastatic prostate cancer who received radiotherapy.

“Different treatment modalities and disease stages are associated with substantial selection bias that may predispose results to false associations,” noted Dr. Deka and coauthors.

Their observational cohort study comprised 45,218 men diagnosed with nonmetastatic prostate cancer at the U.S. Department of Veterans Affairs who underwent radiotherapy with or without ADT. The investigators excluded men who had a diagnosis of dementia within 1 year of the prostate cancer diagnosis or who had prior diagnoses of dementia, stroke, or cognitive impairment.

A total of 1,497 patients were diagnosed with dementia over a median of 6.8 years of follow-up: 404 with Alzheimer disease, 335 with vascular dementia, and 758 with other types or unclassified dementias.

The investigators found no significant association between use of ADT and development of any dementia, the primary outcome of the analysis (subdistribution hazard ratio [SHR], 1.04; 95% confidence interval, 0.94-1.16; P = .43).

Likewise, there was no association between ADT and vascular dementia, specifically, with an SHR of 1.20 (95% CI, 0.97-1.50; P = .10) or Alzheimer’s disease, with an SHR of 1.11 (95% CI, 0.91-1.36; P = .29).

Duration of ADT longer than 1 year was not significantly associated with dementia, nor was duration shorter than 1 year, with SHRs, of 1.08 and 1.01 respectively, the analysis shows.

The SHRs in these and other analysis reported ranged from 1.00 to 1.21. That is substantially lower than hazard ratios of 1.66 to 2.32 in one previous study linking ADT to dementia, according to the investigators, suggesting that the results of the current analysis were not due to inadequate power to detect differences.

Nevertheless, the findings may not be generalizable to some other populations, they cautioned, since it was focused demographically on veterans, and was limited to radiotherapy-treated patients.

Dr. Deka and coauthors reported no conflict of interest. Their study was funded by grants from the University of California San Diego Center for Precision Radiation Medicine.

SOURCE: Deka R et al. JAMA Oncol. 2018 Oct 11. doi: 10.1001/jamaoncol.2018.4423.

Publications
Topics
Sections

In contrast to other recent studies, androgen deprivation therapy (ADT) had no link to dementia in a observational cohort study of more than 45,000 men with prostate cancer who received definitive radiotherapy, investigators have reported.

No significant associations were found between ADT and Alzheimer’s disease or vascular dementia, or between shorter or longer courses of ADT and any dementia studied, according to Rishi Deka, PhD, of Veterans Affairs San Diego Health Care System, La Jolla, Calif., and coinvestigators.

“These results may mitigate concerns regarding the long-term risks of ADT on cognitive health in the treatment of prostate cancer,” Dr. Deka and colleagues wrote in JAMA Oncology.

Two other recent studies showed strong, statistically significant associations between ADT and dementia in prostate cancer. However, those studies combined patients with local and metastatic disease, receiving ADT in the upfront or recurrent settings, while the present study looked specifically at men with nonmetastatic prostate cancer who received radiotherapy.

“Different treatment modalities and disease stages are associated with substantial selection bias that may predispose results to false associations,” noted Dr. Deka and coauthors.

Their observational cohort study comprised 45,218 men diagnosed with nonmetastatic prostate cancer at the U.S. Department of Veterans Affairs who underwent radiotherapy with or without ADT. The investigators excluded men who had a diagnosis of dementia within 1 year of the prostate cancer diagnosis or who had prior diagnoses of dementia, stroke, or cognitive impairment.

A total of 1,497 patients were diagnosed with dementia over a median of 6.8 years of follow-up: 404 with Alzheimer disease, 335 with vascular dementia, and 758 with other types or unclassified dementias.

The investigators found no significant association between use of ADT and development of any dementia, the primary outcome of the analysis (subdistribution hazard ratio [SHR], 1.04; 95% confidence interval, 0.94-1.16; P = .43).

Likewise, there was no association between ADT and vascular dementia, specifically, with an SHR of 1.20 (95% CI, 0.97-1.50; P = .10) or Alzheimer’s disease, with an SHR of 1.11 (95% CI, 0.91-1.36; P = .29).

Duration of ADT longer than 1 year was not significantly associated with dementia, nor was duration shorter than 1 year, with SHRs, of 1.08 and 1.01 respectively, the analysis shows.

The SHRs in these and other analysis reported ranged from 1.00 to 1.21. That is substantially lower than hazard ratios of 1.66 to 2.32 in one previous study linking ADT to dementia, according to the investigators, suggesting that the results of the current analysis were not due to inadequate power to detect differences.

Nevertheless, the findings may not be generalizable to some other populations, they cautioned, since it was focused demographically on veterans, and was limited to radiotherapy-treated patients.

Dr. Deka and coauthors reported no conflict of interest. Their study was funded by grants from the University of California San Diego Center for Precision Radiation Medicine.

SOURCE: Deka R et al. JAMA Oncol. 2018 Oct 11. doi: 10.1001/jamaoncol.2018.4423.

In contrast to other recent studies, androgen deprivation therapy (ADT) had no link to dementia in a observational cohort study of more than 45,000 men with prostate cancer who received definitive radiotherapy, investigators have reported.

No significant associations were found between ADT and Alzheimer’s disease or vascular dementia, or between shorter or longer courses of ADT and any dementia studied, according to Rishi Deka, PhD, of Veterans Affairs San Diego Health Care System, La Jolla, Calif., and coinvestigators.

“These results may mitigate concerns regarding the long-term risks of ADT on cognitive health in the treatment of prostate cancer,” Dr. Deka and colleagues wrote in JAMA Oncology.

Two other recent studies showed strong, statistically significant associations between ADT and dementia in prostate cancer. However, those studies combined patients with local and metastatic disease, receiving ADT in the upfront or recurrent settings, while the present study looked specifically at men with nonmetastatic prostate cancer who received radiotherapy.

“Different treatment modalities and disease stages are associated with substantial selection bias that may predispose results to false associations,” noted Dr. Deka and coauthors.

Their observational cohort study comprised 45,218 men diagnosed with nonmetastatic prostate cancer at the U.S. Department of Veterans Affairs who underwent radiotherapy with or without ADT. The investigators excluded men who had a diagnosis of dementia within 1 year of the prostate cancer diagnosis or who had prior diagnoses of dementia, stroke, or cognitive impairment.

A total of 1,497 patients were diagnosed with dementia over a median of 6.8 years of follow-up: 404 with Alzheimer disease, 335 with vascular dementia, and 758 with other types or unclassified dementias.

The investigators found no significant association between use of ADT and development of any dementia, the primary outcome of the analysis (subdistribution hazard ratio [SHR], 1.04; 95% confidence interval, 0.94-1.16; P = .43).

Likewise, there was no association between ADT and vascular dementia, specifically, with an SHR of 1.20 (95% CI, 0.97-1.50; P = .10) or Alzheimer’s disease, with an SHR of 1.11 (95% CI, 0.91-1.36; P = .29).

Duration of ADT longer than 1 year was not significantly associated with dementia, nor was duration shorter than 1 year, with SHRs, of 1.08 and 1.01 respectively, the analysis shows.

The SHRs in these and other analysis reported ranged from 1.00 to 1.21. That is substantially lower than hazard ratios of 1.66 to 2.32 in one previous study linking ADT to dementia, according to the investigators, suggesting that the results of the current analysis were not due to inadequate power to detect differences.

Nevertheless, the findings may not be generalizable to some other populations, they cautioned, since it was focused demographically on veterans, and was limited to radiotherapy-treated patients.

Dr. Deka and coauthors reported no conflict of interest. Their study was funded by grants from the University of California San Diego Center for Precision Radiation Medicine.

SOURCE: Deka R et al. JAMA Oncol. 2018 Oct 11. doi: 10.1001/jamaoncol.2018.4423.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM JAMA ONCOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

Key clinical point: In contrast with other recent investigations in prostate cancer, researchers found no link between androgen deprivation therapy (ADT) and development of dementia.

Major finding: No significant association was found between use of ADT and development of any dementia (subdistribution hazard ratio [SHR], 1.04; 95% CI, 0.94-1.16; P = .43).

Study details: Observational cohort study of more than 45,000 veterans with nonmetastatic prostate cancer treated with radiotherapy with or without ADT.

Disclosures: This study was funded by grants from the University of California San Diego Center for Precision Radiation Medicine. Dr. Deka and coauthors reported no conflict of interest disclosures related to the work.

Source: Deka R et al. JAMA Oncol. 2018 Oct 11. doi: 10.1001/jamaoncol.2018.4423.

Disqus Comments
Default
Use ProPublica

RCC incidence held steady through 2015 while mortality rates plummeted

Article Type
Changed
Fri, 01/18/2019 - 18:02

 

Renal cell carcinoma (RCC) incidence rates increased in recent years, but have stabilized, while mortality rates have dropped precipitously, according to an analysis of more than 20 years of U.S. cancer registry data.

The introduction of antiangiogenic agents is likely one key factor that led to the mortality decrease, said authors of the analysis of Surveillance, Epidemiology, and End Results data from 1992 to 2015.

Incidence trends are likely more complex and may reflect the interplay between increased detection, on one hand, and decreases in modifiable risk factors such as smoking on the other, the authors reported in Clinical Genitourinary Cancer.

The analysis, conducted by Anas M. Saad, a final-year medical student at Ain Shams University, Cairo; Thai H. Ho, MD, PhD, of Mayo Clinic Cancer Center, Phoenix; and coinvestigators, included a total of 104,584 patients with an RCC diagnosis, of whom nearly 64% were male and 80% were white. The majority of tumors were small and localized at diagnosis, and clear cell was the histologic subtype in 44%, according to the report.

Overall incidence of RCC was 11.3 per 100,000 person-years over the 1992-2015 study period, Dr. Saad and coauthors said in their report.

The incidence rate increased by about 2.4% per year, averaged over the course of the entire study period, though the plateau in rates began around 2008, according to the investigators. A figure in their report shows that the age-adjusted rate was just over 8 per 100,000 person-years in 1992; it climbed steadily until 2008, at which point it remained in the range of about 12-14 per 100,000 person-years for the next 7 years.

The uptick in incidence from 1992 to 2008 was concentrated mostly in localized and regional RCC, rather than distant disease, according to Dr. Saad and colleagues.

The overall incidence-based mortality rate for RCC was 5.3 per 100,000 person-years from 1992 to 2015, Dr. Saad and coauthors said.

Mortality rates increased from 1992 and peaked in 2001, at which point they started to drop at an ever accelerating pace. The annual percent decrease in that mortality rate was 1.5% between 2001 and 2008, 9.3% between 2008 and 2013, and 32.2% from 2013 to 2015, according to the report.

Incidence rate trends are probably affected by increases in incidental diagnoses and changes in RCC risk factor prevalence, investigators noted. For example, there has been a significant increase in use of advanced abdominal imaging, which has improved sensitivity in picking up renal masses, but cannot reliably distinguish between benign and malignant features, they said. On the other hand, smoking, which increases risk of RCC, has been trending downward for decades, which they said correlated with RCC trends.

Authors said RCC survival has been improved by antiangiogenic agents known as vascular endothelial growth factor inhibitors, and more recently immune checkpoint therapies, as clinical trials have shown.

“The decreasing mortality trend starting in 2007 and continuing until 2015 is associated with the introduction of such therapies for RCC treatment,” Dr. Saad and coauthors said in their report.

Support for the study came from the National Cancer Institute and the Department of Defense. Dr. Saad and coauthors declared that they had no conflicts of interest.

SOURCE: Saad AM et al. Clin Genitourin Cancer. 2018. doi: 10.1016/j.clgc.2018.10.002.

Publications
Topics
Sections

 

Renal cell carcinoma (RCC) incidence rates increased in recent years, but have stabilized, while mortality rates have dropped precipitously, according to an analysis of more than 20 years of U.S. cancer registry data.

The introduction of antiangiogenic agents is likely one key factor that led to the mortality decrease, said authors of the analysis of Surveillance, Epidemiology, and End Results data from 1992 to 2015.

Incidence trends are likely more complex and may reflect the interplay between increased detection, on one hand, and decreases in modifiable risk factors such as smoking on the other, the authors reported in Clinical Genitourinary Cancer.

The analysis, conducted by Anas M. Saad, a final-year medical student at Ain Shams University, Cairo; Thai H. Ho, MD, PhD, of Mayo Clinic Cancer Center, Phoenix; and coinvestigators, included a total of 104,584 patients with an RCC diagnosis, of whom nearly 64% were male and 80% were white. The majority of tumors were small and localized at diagnosis, and clear cell was the histologic subtype in 44%, according to the report.

Overall incidence of RCC was 11.3 per 100,000 person-years over the 1992-2015 study period, Dr. Saad and coauthors said in their report.

The incidence rate increased by about 2.4% per year, averaged over the course of the entire study period, though the plateau in rates began around 2008, according to the investigators. A figure in their report shows that the age-adjusted rate was just over 8 per 100,000 person-years in 1992; it climbed steadily until 2008, at which point it remained in the range of about 12-14 per 100,000 person-years for the next 7 years.

The uptick in incidence from 1992 to 2008 was concentrated mostly in localized and regional RCC, rather than distant disease, according to Dr. Saad and colleagues.

The overall incidence-based mortality rate for RCC was 5.3 per 100,000 person-years from 1992 to 2015, Dr. Saad and coauthors said.

Mortality rates increased from 1992 and peaked in 2001, at which point they started to drop at an ever accelerating pace. The annual percent decrease in that mortality rate was 1.5% between 2001 and 2008, 9.3% between 2008 and 2013, and 32.2% from 2013 to 2015, according to the report.

Incidence rate trends are probably affected by increases in incidental diagnoses and changes in RCC risk factor prevalence, investigators noted. For example, there has been a significant increase in use of advanced abdominal imaging, which has improved sensitivity in picking up renal masses, but cannot reliably distinguish between benign and malignant features, they said. On the other hand, smoking, which increases risk of RCC, has been trending downward for decades, which they said correlated with RCC trends.

Authors said RCC survival has been improved by antiangiogenic agents known as vascular endothelial growth factor inhibitors, and more recently immune checkpoint therapies, as clinical trials have shown.

“The decreasing mortality trend starting in 2007 and continuing until 2015 is associated with the introduction of such therapies for RCC treatment,” Dr. Saad and coauthors said in their report.

Support for the study came from the National Cancer Institute and the Department of Defense. Dr. Saad and coauthors declared that they had no conflicts of interest.

SOURCE: Saad AM et al. Clin Genitourin Cancer. 2018. doi: 10.1016/j.clgc.2018.10.002.

 

Renal cell carcinoma (RCC) incidence rates increased in recent years, but have stabilized, while mortality rates have dropped precipitously, according to an analysis of more than 20 years of U.S. cancer registry data.

The introduction of antiangiogenic agents is likely one key factor that led to the mortality decrease, said authors of the analysis of Surveillance, Epidemiology, and End Results data from 1992 to 2015.

Incidence trends are likely more complex and may reflect the interplay between increased detection, on one hand, and decreases in modifiable risk factors such as smoking on the other, the authors reported in Clinical Genitourinary Cancer.

The analysis, conducted by Anas M. Saad, a final-year medical student at Ain Shams University, Cairo; Thai H. Ho, MD, PhD, of Mayo Clinic Cancer Center, Phoenix; and coinvestigators, included a total of 104,584 patients with an RCC diagnosis, of whom nearly 64% were male and 80% were white. The majority of tumors were small and localized at diagnosis, and clear cell was the histologic subtype in 44%, according to the report.

Overall incidence of RCC was 11.3 per 100,000 person-years over the 1992-2015 study period, Dr. Saad and coauthors said in their report.

The incidence rate increased by about 2.4% per year, averaged over the course of the entire study period, though the plateau in rates began around 2008, according to the investigators. A figure in their report shows that the age-adjusted rate was just over 8 per 100,000 person-years in 1992; it climbed steadily until 2008, at which point it remained in the range of about 12-14 per 100,000 person-years for the next 7 years.

The uptick in incidence from 1992 to 2008 was concentrated mostly in localized and regional RCC, rather than distant disease, according to Dr. Saad and colleagues.

The overall incidence-based mortality rate for RCC was 5.3 per 100,000 person-years from 1992 to 2015, Dr. Saad and coauthors said.

Mortality rates increased from 1992 and peaked in 2001, at which point they started to drop at an ever accelerating pace. The annual percent decrease in that mortality rate was 1.5% between 2001 and 2008, 9.3% between 2008 and 2013, and 32.2% from 2013 to 2015, according to the report.

Incidence rate trends are probably affected by increases in incidental diagnoses and changes in RCC risk factor prevalence, investigators noted. For example, there has been a significant increase in use of advanced abdominal imaging, which has improved sensitivity in picking up renal masses, but cannot reliably distinguish between benign and malignant features, they said. On the other hand, smoking, which increases risk of RCC, has been trending downward for decades, which they said correlated with RCC trends.

Authors said RCC survival has been improved by antiangiogenic agents known as vascular endothelial growth factor inhibitors, and more recently immune checkpoint therapies, as clinical trials have shown.

“The decreasing mortality trend starting in 2007 and continuing until 2015 is associated with the introduction of such therapies for RCC treatment,” Dr. Saad and coauthors said in their report.

Support for the study came from the National Cancer Institute and the Department of Defense. Dr. Saad and coauthors declared that they had no conflicts of interest.

SOURCE: Saad AM et al. Clin Genitourin Cancer. 2018. doi: 10.1016/j.clgc.2018.10.002.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CLINICAL GENITOURINARY CANCER

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: In recent years, renal cell carcinoma incidence rates have stabilized, while mortality due to the disease has dropped sharply.

Major finding: Mortality rates increased from 1992 and peaked in 2001, at which point they started to drop at an ever accelerating pace, culminating in an annual percent decrease of 32.2% in the 2013-2015 time period.

Study details: Analysis including more than 104,000 patients with an RCC diagnosis in the Surveillance, Epidemiology and End Results (SEER) database from 1992 to 2015.

Disclosures: Support for the study came from the National Cancer Institute and the Department of Defense. Study authors declared that they had no conflicts of interest.

Source: Saad AM et al. Clin Genitourin Cancer. 2018. doi: 10.1016/j.clgc.2018.10.002.

Disqus Comments
Default
Use ProPublica

Poor outcomes for RCC with nodal involvement: Time to reconsider staging criteria?

Article Type
Changed
Fri, 01/04/2019 - 14:25

 

Among patients with stage III renal cell carcinoma (RCC) by current staging criteria, survival outcomes are worse for those who have pathological nodal involvement, results of a retrospective study suggest.

Survival was significantly shorter for stage III patients with nodal involvement versus those with no involvement, and was “equivalent” to survival in stage IV RCC patients, according to study author Jose A. Karam, MD, of the University of Texas MD Anderson Cancer Center, Houston, and his coauthors.

“If these findings are validated in other studies, consideration should be given to reclassifying patients with stage III, pN1 disease as having stage IV disease,” Dr. Karam and his coauthors wrote in Cancer.

Dr. Karam and his colleagues looked at records for patients with RCC of any histologic subtype who had undergone radical or partial nephrectomy between 1993 and 2012. Their analysis included a total of 389 patients with stage III disease, including 274 with node-positive disease (pT3N0M0) and 115 with node-negative disease (pT123N1M0), along with 523 patients who had stage IV disease.

They found that median overall survival was just 2.4 years (95% confidence interval, 1.7-4.1) for stage III, node-positive disease patients, compared with 10.2 years (95% CI, 8.7-not estimable [NE]) for stage III, node-negative disease and 2.4 years (95% CI, 2.1-3.0) for stage IV disease.

There was a significant difference in overall survival between the three groups, but no significant difference between patients with stage III node-positive disease and stage IV disease, the investigators wrote.

Similarly, median cancer-specific survival was 2.8 years (95% CI, 1.8-4.8) for stage III node-positive disease, not reached (95% CI, 10.2-NE) for stage III node-negative disease, and 2.4 years (95% CI, 2.1-3.0) for stage IV disease, the investigators wrote.

In multivariate analysis, pathological lymph node involvement in the stage III patients was independently associated with worse overall and cancer-specific survival.

Dr. Karam and his coauthors wrote that it may be prudent to revise the current staging system to reclassify node-positive patients if further research confirms their findings. By the current American Joint Committee on Cancer tumor, nodes, and metastases staging manual, patients can be classified as stage III on the basis of either primary tumor status (pT3) or pathological lymph node involvement, they said.

The incidence of pathological, node-positive disease in RCC ranges from 2%-10% in studies, with 5-year survival rates that are “poor” at 5%-30%, the authors noted in their report.

“Even in the targeted therapy era, adjuvant therapy with tyrosine kinase inhibitors does not appear to improve on these outcomes in patients with pN-positive disease,” they wrote.

Funding support for the research came from the National Institutes of Health/National Cancer Institute. Dr. Karam reported serving as a consultant/advisory board member for Pfizer, EMD Serono, Novartis, and Roche/Genentech, and that the MD Anderson Cancer Center has received clinical trial research funding from Roche/Genentech, though none of these disclosures were related to the current report.

SOURCE: Karam JA et al. Cancer. 2018 Oct 1. doi: 10.1002/cncr.31661.

Publications
Topics
Sections

 

Among patients with stage III renal cell carcinoma (RCC) by current staging criteria, survival outcomes are worse for those who have pathological nodal involvement, results of a retrospective study suggest.

Survival was significantly shorter for stage III patients with nodal involvement versus those with no involvement, and was “equivalent” to survival in stage IV RCC patients, according to study author Jose A. Karam, MD, of the University of Texas MD Anderson Cancer Center, Houston, and his coauthors.

“If these findings are validated in other studies, consideration should be given to reclassifying patients with stage III, pN1 disease as having stage IV disease,” Dr. Karam and his coauthors wrote in Cancer.

Dr. Karam and his colleagues looked at records for patients with RCC of any histologic subtype who had undergone radical or partial nephrectomy between 1993 and 2012. Their analysis included a total of 389 patients with stage III disease, including 274 with node-positive disease (pT3N0M0) and 115 with node-negative disease (pT123N1M0), along with 523 patients who had stage IV disease.

They found that median overall survival was just 2.4 years (95% confidence interval, 1.7-4.1) for stage III, node-positive disease patients, compared with 10.2 years (95% CI, 8.7-not estimable [NE]) for stage III, node-negative disease and 2.4 years (95% CI, 2.1-3.0) for stage IV disease.

There was a significant difference in overall survival between the three groups, but no significant difference between patients with stage III node-positive disease and stage IV disease, the investigators wrote.

Similarly, median cancer-specific survival was 2.8 years (95% CI, 1.8-4.8) for stage III node-positive disease, not reached (95% CI, 10.2-NE) for stage III node-negative disease, and 2.4 years (95% CI, 2.1-3.0) for stage IV disease, the investigators wrote.

In multivariate analysis, pathological lymph node involvement in the stage III patients was independently associated with worse overall and cancer-specific survival.

Dr. Karam and his coauthors wrote that it may be prudent to revise the current staging system to reclassify node-positive patients if further research confirms their findings. By the current American Joint Committee on Cancer tumor, nodes, and metastases staging manual, patients can be classified as stage III on the basis of either primary tumor status (pT3) or pathological lymph node involvement, they said.

The incidence of pathological, node-positive disease in RCC ranges from 2%-10% in studies, with 5-year survival rates that are “poor” at 5%-30%, the authors noted in their report.

“Even in the targeted therapy era, adjuvant therapy with tyrosine kinase inhibitors does not appear to improve on these outcomes in patients with pN-positive disease,” they wrote.

Funding support for the research came from the National Institutes of Health/National Cancer Institute. Dr. Karam reported serving as a consultant/advisory board member for Pfizer, EMD Serono, Novartis, and Roche/Genentech, and that the MD Anderson Cancer Center has received clinical trial research funding from Roche/Genentech, though none of these disclosures were related to the current report.

SOURCE: Karam JA et al. Cancer. 2018 Oct 1. doi: 10.1002/cncr.31661.

 

Among patients with stage III renal cell carcinoma (RCC) by current staging criteria, survival outcomes are worse for those who have pathological nodal involvement, results of a retrospective study suggest.

Survival was significantly shorter for stage III patients with nodal involvement versus those with no involvement, and was “equivalent” to survival in stage IV RCC patients, according to study author Jose A. Karam, MD, of the University of Texas MD Anderson Cancer Center, Houston, and his coauthors.

“If these findings are validated in other studies, consideration should be given to reclassifying patients with stage III, pN1 disease as having stage IV disease,” Dr. Karam and his coauthors wrote in Cancer.

Dr. Karam and his colleagues looked at records for patients with RCC of any histologic subtype who had undergone radical or partial nephrectomy between 1993 and 2012. Their analysis included a total of 389 patients with stage III disease, including 274 with node-positive disease (pT3N0M0) and 115 with node-negative disease (pT123N1M0), along with 523 patients who had stage IV disease.

They found that median overall survival was just 2.4 years (95% confidence interval, 1.7-4.1) for stage III, node-positive disease patients, compared with 10.2 years (95% CI, 8.7-not estimable [NE]) for stage III, node-negative disease and 2.4 years (95% CI, 2.1-3.0) for stage IV disease.

There was a significant difference in overall survival between the three groups, but no significant difference between patients with stage III node-positive disease and stage IV disease, the investigators wrote.

Similarly, median cancer-specific survival was 2.8 years (95% CI, 1.8-4.8) for stage III node-positive disease, not reached (95% CI, 10.2-NE) for stage III node-negative disease, and 2.4 years (95% CI, 2.1-3.0) for stage IV disease, the investigators wrote.

In multivariate analysis, pathological lymph node involvement in the stage III patients was independently associated with worse overall and cancer-specific survival.

Dr. Karam and his coauthors wrote that it may be prudent to revise the current staging system to reclassify node-positive patients if further research confirms their findings. By the current American Joint Committee on Cancer tumor, nodes, and metastases staging manual, patients can be classified as stage III on the basis of either primary tumor status (pT3) or pathological lymph node involvement, they said.

The incidence of pathological, node-positive disease in RCC ranges from 2%-10% in studies, with 5-year survival rates that are “poor” at 5%-30%, the authors noted in their report.

“Even in the targeted therapy era, adjuvant therapy with tyrosine kinase inhibitors does not appear to improve on these outcomes in patients with pN-positive disease,” they wrote.

Funding support for the research came from the National Institutes of Health/National Cancer Institute. Dr. Karam reported serving as a consultant/advisory board member for Pfizer, EMD Serono, Novartis, and Roche/Genentech, and that the MD Anderson Cancer Center has received clinical trial research funding from Roche/Genentech, though none of these disclosures were related to the current report.

SOURCE: Karam JA et al. Cancer. 2018 Oct 1. doi: 10.1002/cncr.31661.

Publications
Publications
Topics
Article Type
Click for Credit Status
Active
Sections
Article Source

FROM CANCER

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
CME ID
177193
Vitals

 

Key clinical point: Among patients with stage III renal cell carcinoma, survival outcomes for those with nodal involvement are worse than for those with no nodal involvement and comparable with patients with stage IV disease, suggesting a potential need to revisit current staging criteria.

Major finding: Overall survival was 2.4 years for stage III node-positive disease, 10.2 years for stage III, node-negative disease, and 2.4 years for stage IV disease.

Study details: A retrospective study including 912 patients with renal cell carcinoma who underwent radical or partial nephrectomy between 1993 and 2012.

Disclosures: Funding was reported from the National Institutes of Health/National Cancer Institute. One study author provided disclosures with Pfizer, EMD Serono, Novartis, and Roche/Genentech.

Source: Karam JA et al. Cancer. 2018 Oct 1. doi: 10.1002/cncr.31661.

Disqus Comments
Default
Use ProPublica

Primary, metastatic ccRCC share mutational features

Article Type
Changed
Fri, 01/04/2019 - 14:25

 

A comparison of the genomic landscapes of both primary and metastatic clear cell renal cell carcinoma (ccRCC) found no significant differences in gene expression or mutational burden between the disease states, suggesting that there is no single genetic driver of metastases, investigators reported.

An analysis of targeted next-generation sequencing of both primary tumors and metastases in two independent patient cohorts showed that only the gene encoding for the tumor suppressor TP53 was significantly more frequently mutated in metastases, compared with primary tumors, but this finding did not pass a false positive test (false discovery rate), noted Toni K. Choueiri, MD, from the Dana-Farber Cancer Institute in Boston, and his colleagues, in the British Journal of Cancer.

“No other gene had significant difference in the cohort frequency of mutations between the metastases and primary tumors. Mutation burden was not significantly different between the metastases and primary tumors or between metastatic sites,” they wrote.

Frequently mutated genes in ccRCC include VHL, the gene encoding for von Hippel–Lindau syndrome, as well as tumor suppressor genes such as PBRM1, SEDT2, BAP1 and KDM5C, but few mutations are clinically actionable, the investigators noted. “However, the value of genomic alterations will be determined by understanding the interactions between acquired genetic alterations, treatments received, heterogeneity, and the dynamics of mutations during evolution of disease.”

To see whether they could improve understanding of the genomic differences between primary and metastatic ccRCC and potentially develop personalized therapies, the investigators studied targeted next-generation sequencing data from two separate cohorts.

The first cohort included data on 349 ccRCC primary tumors and 229 unmatched cases of metastatic ccRCC from the Foundation Medicine database. The second, a validation cohort, included data on 177 ccRCC primary tumors and 80 metastases from patients treated at Dana-Farber. In each cohort, sequencing was performed on 275 genes and intronic regions in 30 genes for a total of 282 unique genes.

In cohort 1, which included tumor samples from 417 men and 169 women with a median age of 58 years, the frequency of mutations was similar in primary tumors and metastases. As noted before, mutations in TP53 were significantly more frequent in metastases than in primary tumors, detected in 14.85% versus 8.90% of samples, respectively (P = .031). This difference did not, however, pass the false discovery rate test (q = 0.21). Two other tumor suppressor genes, PBRM1 and KDM5C, were numerically but not significantly more frequent in metastases.

There were no differences in median tumor mutational burden between primary and metastatic samples and no difference in either mutations or mutational burden across different metastatic sites.

In cohort 2 there were no significantly different mutational frequencies between primary and metastatic samples for any gene and no differences in median tumor mutational burden.

An analysis comparing the frequency of gene mutations in primary tumors of patients in this cohort who went on to develop metastatic disease versus those of patients with only localized disease showed that, after a median follow-up of 21.9 months, there were no significant differences in either mutational frequency or tumor mutational burden.

“It is currently unknown if cohortwide genomic alterations in RCC metastases have a different genomic profile, including potential actionable mutations, compared to samples derived from the primary site. To our knowledge, our analysis is the largest genomic ccRCC study that compares cohortwide mutational differences between metastases and primary tumors,” the investigators wrote.

They acknowledged that because they did not match primary tumors with metastatic tumors in the same patients they were unable to evaluate how individual tumors evolve over time or how systemic therapies may alter the tumor genomic landscape.

Dr. Choueri is supported in part by the Dana-Farber/Harvard Cancer Center Kidney SPORE, the Kohlberg Chair at Harvard Medical School and the Trust Family, Michael Brigham, and Loker Pinard Funds for Kidney Cancer Research at the Dana-Farber Cancer Institute. Six of the study’s coauthors are employed by Foundation Medicine. The remaining authors declared no competing interests.

SOURCE: Choueiri TK et al. Br J Cancer. 2018 May;118(9):1238-42.

Publications
Topics
Sections

 

A comparison of the genomic landscapes of both primary and metastatic clear cell renal cell carcinoma (ccRCC) found no significant differences in gene expression or mutational burden between the disease states, suggesting that there is no single genetic driver of metastases, investigators reported.

An analysis of targeted next-generation sequencing of both primary tumors and metastases in two independent patient cohorts showed that only the gene encoding for the tumor suppressor TP53 was significantly more frequently mutated in metastases, compared with primary tumors, but this finding did not pass a false positive test (false discovery rate), noted Toni K. Choueiri, MD, from the Dana-Farber Cancer Institute in Boston, and his colleagues, in the British Journal of Cancer.

“No other gene had significant difference in the cohort frequency of mutations between the metastases and primary tumors. Mutation burden was not significantly different between the metastases and primary tumors or between metastatic sites,” they wrote.

Frequently mutated genes in ccRCC include VHL, the gene encoding for von Hippel–Lindau syndrome, as well as tumor suppressor genes such as PBRM1, SEDT2, BAP1 and KDM5C, but few mutations are clinically actionable, the investigators noted. “However, the value of genomic alterations will be determined by understanding the interactions between acquired genetic alterations, treatments received, heterogeneity, and the dynamics of mutations during evolution of disease.”

To see whether they could improve understanding of the genomic differences between primary and metastatic ccRCC and potentially develop personalized therapies, the investigators studied targeted next-generation sequencing data from two separate cohorts.

The first cohort included data on 349 ccRCC primary tumors and 229 unmatched cases of metastatic ccRCC from the Foundation Medicine database. The second, a validation cohort, included data on 177 ccRCC primary tumors and 80 metastases from patients treated at Dana-Farber. In each cohort, sequencing was performed on 275 genes and intronic regions in 30 genes for a total of 282 unique genes.

In cohort 1, which included tumor samples from 417 men and 169 women with a median age of 58 years, the frequency of mutations was similar in primary tumors and metastases. As noted before, mutations in TP53 were significantly more frequent in metastases than in primary tumors, detected in 14.85% versus 8.90% of samples, respectively (P = .031). This difference did not, however, pass the false discovery rate test (q = 0.21). Two other tumor suppressor genes, PBRM1 and KDM5C, were numerically but not significantly more frequent in metastases.

There were no differences in median tumor mutational burden between primary and metastatic samples and no difference in either mutations or mutational burden across different metastatic sites.

In cohort 2 there were no significantly different mutational frequencies between primary and metastatic samples for any gene and no differences in median tumor mutational burden.

An analysis comparing the frequency of gene mutations in primary tumors of patients in this cohort who went on to develop metastatic disease versus those of patients with only localized disease showed that, after a median follow-up of 21.9 months, there were no significant differences in either mutational frequency or tumor mutational burden.

“It is currently unknown if cohortwide genomic alterations in RCC metastases have a different genomic profile, including potential actionable mutations, compared to samples derived from the primary site. To our knowledge, our analysis is the largest genomic ccRCC study that compares cohortwide mutational differences between metastases and primary tumors,” the investigators wrote.

They acknowledged that because they did not match primary tumors with metastatic tumors in the same patients they were unable to evaluate how individual tumors evolve over time or how systemic therapies may alter the tumor genomic landscape.

Dr. Choueri is supported in part by the Dana-Farber/Harvard Cancer Center Kidney SPORE, the Kohlberg Chair at Harvard Medical School and the Trust Family, Michael Brigham, and Loker Pinard Funds for Kidney Cancer Research at the Dana-Farber Cancer Institute. Six of the study’s coauthors are employed by Foundation Medicine. The remaining authors declared no competing interests.

SOURCE: Choueiri TK et al. Br J Cancer. 2018 May;118(9):1238-42.

 

A comparison of the genomic landscapes of both primary and metastatic clear cell renal cell carcinoma (ccRCC) found no significant differences in gene expression or mutational burden between the disease states, suggesting that there is no single genetic driver of metastases, investigators reported.

An analysis of targeted next-generation sequencing of both primary tumors and metastases in two independent patient cohorts showed that only the gene encoding for the tumor suppressor TP53 was significantly more frequently mutated in metastases, compared with primary tumors, but this finding did not pass a false positive test (false discovery rate), noted Toni K. Choueiri, MD, from the Dana-Farber Cancer Institute in Boston, and his colleagues, in the British Journal of Cancer.

“No other gene had significant difference in the cohort frequency of mutations between the metastases and primary tumors. Mutation burden was not significantly different between the metastases and primary tumors or between metastatic sites,” they wrote.

Frequently mutated genes in ccRCC include VHL, the gene encoding for von Hippel–Lindau syndrome, as well as tumor suppressor genes such as PBRM1, SEDT2, BAP1 and KDM5C, but few mutations are clinically actionable, the investigators noted. “However, the value of genomic alterations will be determined by understanding the interactions between acquired genetic alterations, treatments received, heterogeneity, and the dynamics of mutations during evolution of disease.”

To see whether they could improve understanding of the genomic differences between primary and metastatic ccRCC and potentially develop personalized therapies, the investigators studied targeted next-generation sequencing data from two separate cohorts.

The first cohort included data on 349 ccRCC primary tumors and 229 unmatched cases of metastatic ccRCC from the Foundation Medicine database. The second, a validation cohort, included data on 177 ccRCC primary tumors and 80 metastases from patients treated at Dana-Farber. In each cohort, sequencing was performed on 275 genes and intronic regions in 30 genes for a total of 282 unique genes.

In cohort 1, which included tumor samples from 417 men and 169 women with a median age of 58 years, the frequency of mutations was similar in primary tumors and metastases. As noted before, mutations in TP53 were significantly more frequent in metastases than in primary tumors, detected in 14.85% versus 8.90% of samples, respectively (P = .031). This difference did not, however, pass the false discovery rate test (q = 0.21). Two other tumor suppressor genes, PBRM1 and KDM5C, were numerically but not significantly more frequent in metastases.

There were no differences in median tumor mutational burden between primary and metastatic samples and no difference in either mutations or mutational burden across different metastatic sites.

In cohort 2 there were no significantly different mutational frequencies between primary and metastatic samples for any gene and no differences in median tumor mutational burden.

An analysis comparing the frequency of gene mutations in primary tumors of patients in this cohort who went on to develop metastatic disease versus those of patients with only localized disease showed that, after a median follow-up of 21.9 months, there were no significant differences in either mutational frequency or tumor mutational burden.

“It is currently unknown if cohortwide genomic alterations in RCC metastases have a different genomic profile, including potential actionable mutations, compared to samples derived from the primary site. To our knowledge, our analysis is the largest genomic ccRCC study that compares cohortwide mutational differences between metastases and primary tumors,” the investigators wrote.

They acknowledged that because they did not match primary tumors with metastatic tumors in the same patients they were unable to evaluate how individual tumors evolve over time or how systemic therapies may alter the tumor genomic landscape.

Dr. Choueri is supported in part by the Dana-Farber/Harvard Cancer Center Kidney SPORE, the Kohlberg Chair at Harvard Medical School and the Trust Family, Michael Brigham, and Loker Pinard Funds for Kidney Cancer Research at the Dana-Farber Cancer Institute. Six of the study’s coauthors are employed by Foundation Medicine. The remaining authors declared no competing interests.

SOURCE: Choueiri TK et al. Br J Cancer. 2018 May;118(9):1238-42.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM THE BRITISH JOURNAL OF CANCER

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: There does not appear to be a single-gene driver of metastases in patients with clear cell renal cell carcinoma.

Major finding: There were no significant differences in mutational frequency or tumor mutational burden between primary and metastatic clear cell renal cell carcinoma.

Study details: A targeted next-generation sequencing analysis of data from two independent patient cohorts.

Disclosures: Dr. Choueri is supported in part by the Dana-Farber/Harvard Cancer Center Kidney SPORE, the Kohlberg Chair at Harvard Medical School and the Trust Family, Michael Brigham, and Loker Pinard Funds for Kidney Cancer Research at the Dana-Farber Cancer Institute. Six of the study’s coauthors are employed by Foundation Medicine. The remaining authors reported no competing interests.

Source: Choueiri TK et al. Br J Cancer. 2018 May;118(9):1238-42.

Disqus Comments
Default
Use ProPublica

Updated analysis from JAVELIN Renal 101 to be presented at ESMO 2018

Article Type
Changed
Fri, 01/04/2019 - 14:24

An updated analysis of interim results for JAVELIN Renal 101 will be presented at a Presidential Symposium during the annual congress of the European Society for Medical Oncology (ESMO 2018), to be held Oct. 19-23 in Munich.

The phase 3 trial compared avelumab (Bavencio) in combination with axitinib (Inlyta) to sunitinib monotherapy as first-line treatment, in patients with advanced renal cell carcinoma (RCC). Interim analysis results announced by the company in a September press release indicated the combination of immunotherapy and a tyrosine kinase inhibitor showed “a statistically significant improvement in progression-free survival by central review for patients treated with the combination whose tumors had programmed death ligand-1‒positive (PD-L1+) expression greater than 1% (primary objective), as well as in the entire study population regardless of PD-L1 tumor expression (secondary objective).”

An updated analysis of progression-free survival and overall response rates will be presented at ESMO 2018 by Robert J. Motzer, MD of Memorial Sloan Kettering Cancer Center, New York.

A phase 1b study (JAVELIN Renal 100), published in Lancet Oncology, found the safety profile of the combination to be similar to either drug alone.

For the phase 3 trial, more than 800 patients with advanced RCC were randomized to first-line treatment with the combination of avelumab (10 mg/kg IV every 2 weeks) plus axitinib (5 mg orally, twice daily) or monotherapy with sunitinib (50 mg orally once daily, 4 weeks on/2 weeks off). The overall survival results will be presented at the Presidential Symposium 2 on Oct. 21.

This article was updated on 10/15/18 to reflect the fact that interim results will be presented.






 

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

An updated analysis of interim results for JAVELIN Renal 101 will be presented at a Presidential Symposium during the annual congress of the European Society for Medical Oncology (ESMO 2018), to be held Oct. 19-23 in Munich.

The phase 3 trial compared avelumab (Bavencio) in combination with axitinib (Inlyta) to sunitinib monotherapy as first-line treatment, in patients with advanced renal cell carcinoma (RCC). Interim analysis results announced by the company in a September press release indicated the combination of immunotherapy and a tyrosine kinase inhibitor showed “a statistically significant improvement in progression-free survival by central review for patients treated with the combination whose tumors had programmed death ligand-1‒positive (PD-L1+) expression greater than 1% (primary objective), as well as in the entire study population regardless of PD-L1 tumor expression (secondary objective).”

An updated analysis of progression-free survival and overall response rates will be presented at ESMO 2018 by Robert J. Motzer, MD of Memorial Sloan Kettering Cancer Center, New York.

A phase 1b study (JAVELIN Renal 100), published in Lancet Oncology, found the safety profile of the combination to be similar to either drug alone.

For the phase 3 trial, more than 800 patients with advanced RCC were randomized to first-line treatment with the combination of avelumab (10 mg/kg IV every 2 weeks) plus axitinib (5 mg orally, twice daily) or monotherapy with sunitinib (50 mg orally once daily, 4 weeks on/2 weeks off). The overall survival results will be presented at the Presidential Symposium 2 on Oct. 21.

This article was updated on 10/15/18 to reflect the fact that interim results will be presented.






 

An updated analysis of interim results for JAVELIN Renal 101 will be presented at a Presidential Symposium during the annual congress of the European Society for Medical Oncology (ESMO 2018), to be held Oct. 19-23 in Munich.

The phase 3 trial compared avelumab (Bavencio) in combination with axitinib (Inlyta) to sunitinib monotherapy as first-line treatment, in patients with advanced renal cell carcinoma (RCC). Interim analysis results announced by the company in a September press release indicated the combination of immunotherapy and a tyrosine kinase inhibitor showed “a statistically significant improvement in progression-free survival by central review for patients treated with the combination whose tumors had programmed death ligand-1‒positive (PD-L1+) expression greater than 1% (primary objective), as well as in the entire study population regardless of PD-L1 tumor expression (secondary objective).”

An updated analysis of progression-free survival and overall response rates will be presented at ESMO 2018 by Robert J. Motzer, MD of Memorial Sloan Kettering Cancer Center, New York.

A phase 1b study (JAVELIN Renal 100), published in Lancet Oncology, found the safety profile of the combination to be similar to either drug alone.

For the phase 3 trial, more than 800 patients with advanced RCC were randomized to first-line treatment with the combination of avelumab (10 mg/kg IV every 2 weeks) plus axitinib (5 mg orally, twice daily) or monotherapy with sunitinib (50 mg orally once daily, 4 weeks on/2 weeks off). The overall survival results will be presented at the Presidential Symposium 2 on Oct. 21.

This article was updated on 10/15/18 to reflect the fact that interim results will be presented.






 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica