User login
Active surveillance an option for patients with mRCC
ORLANDO – Active surveillance prior to initiating targeted therapy could be an option for some patients with metastatic renal cell carcinoma (mRCC), according to new findings.
“Active surveillance does not affect the efficacy of subsequent therapies,” said lead study author Davide Bimbatti, MD, of the Azienda Ospedaliera Universitaria Integrata, University of Verona (Italy). “In selected patients, active surveillance allows us to delay the start of systemic treatment, and patients in active surveillance rarely have a worsening of prognostic class,” he said in a press briefing held at the 2017 Genitourinary Cancers Symposium sponsored by the American Society of Clinical Oncology, ASTRO, and the Society of Urologic Oncology.
Targeted therapies can improve survival in mRCC but treatment is not curative and associated toxicities can interfere with quality of life and force patients to discontinue treatment. Active surveillance delays the use of systemic therapy and associated toxicities, and is a feasible strategy for patients with indolent disease.
However, the effect of active surveillance on tumor burden and prognosis have not been investigated.
To address this issue, Dr. Bimbatti and his colleagues conducted a retrospective study and evaluated the effect of active surveillance on overall survival and postsurveillance overall survival, progression-free survival, and tumor burden (a measure of the number of sites and tumor size).
The study cohort included 48 patients with mRCC who underwent active surveillance during 2007-2016 and changes in the International mRCC Database Consortium (IMDC) prognostic class and tumor burden were analyzed for associations with these endpoints.
At baseline, 69% of patients had a favorable prognostic IMDC class, 25% were at an intermediate class, and 6% had a poor class designation. The main sites of metastases were lung (56%), lymph nodes (25%), pancreas (14%), adrenal gland (8%), CNS (8%), and bone (6%).
At a median follow-up of 37.3 months, 79.2% of patients were still alive and, at a median surveillance duration of 16.7 months, 71% of patients had begun a targeted therapy. There were a total of 17 deaths (33%).
Median progression-free survival was 16.6 months and median postsurveillance overall survival was 39.1 months.
During active surveillance, only four patients transitioned from good to intermediate IMDC prognostic class. IMDC classes overall maintained their prognostic value during active surveillance, Dr. Bimbatti said. IMDC class was also the only factor that was associated with time on surveillance.
At baseline, tumor burden was in one site for 65% of patients, two sites in 31%, and three or more sites for 4%, but during active surveillance, changes occurred in one site in 35% of patients, two in 48%, and more than two in 17%.
A change in tumor burden (greater than or equal to 2.2 times the original burden), however, was related to poorer postsurveillance survival (hazard ratio,1.23; P less than .01), but not with overall survival (HR, 1.0; P less than .05).
Conversely, any increase in metastatic sites was associated with both significantly worse postsurveillance and overall survival (HR = 2.6, P = 0.04; and HR = 3.3, P less than 0.01).
Commenting on the study, Primo N. Lara Jr., MD, of the University of California, Davis, Comprehensive Cancer Center explained that active surveillance “remains a reasonable option for highly selected patients.”
“[The investigators] provide reassuring data that active surveillance does not alter the IMDC risk grouping,” he said, and that the results of this study overall, were similar to those of prior research.
There were some limitations to the study, in that it was retrospective and conducted at a single institution. “It also failed to account for psychosocial issues such as anxiety during the active surveillance phase,” he said.
Critical questions also remain, Dr. Lara added, such as what are the validated selection criteria for patients entering active surveillance or what is the threshold of tumor burden to warrant active surveillance discontinuation.
The funding source was not disclosed. Dr. Bimbatti and his coauthors have no disclosures. Dr. Lara reports financial ties to multiple pharmaceutical companies.
ORLANDO – Active surveillance prior to initiating targeted therapy could be an option for some patients with metastatic renal cell carcinoma (mRCC), according to new findings.
“Active surveillance does not affect the efficacy of subsequent therapies,” said lead study author Davide Bimbatti, MD, of the Azienda Ospedaliera Universitaria Integrata, University of Verona (Italy). “In selected patients, active surveillance allows us to delay the start of systemic treatment, and patients in active surveillance rarely have a worsening of prognostic class,” he said in a press briefing held at the 2017 Genitourinary Cancers Symposium sponsored by the American Society of Clinical Oncology, ASTRO, and the Society of Urologic Oncology.
Targeted therapies can improve survival in mRCC but treatment is not curative and associated toxicities can interfere with quality of life and force patients to discontinue treatment. Active surveillance delays the use of systemic therapy and associated toxicities, and is a feasible strategy for patients with indolent disease.
However, the effect of active surveillance on tumor burden and prognosis have not been investigated.
To address this issue, Dr. Bimbatti and his colleagues conducted a retrospective study and evaluated the effect of active surveillance on overall survival and postsurveillance overall survival, progression-free survival, and tumor burden (a measure of the number of sites and tumor size).
The study cohort included 48 patients with mRCC who underwent active surveillance during 2007-2016 and changes in the International mRCC Database Consortium (IMDC) prognostic class and tumor burden were analyzed for associations with these endpoints.
At baseline, 69% of patients had a favorable prognostic IMDC class, 25% were at an intermediate class, and 6% had a poor class designation. The main sites of metastases were lung (56%), lymph nodes (25%), pancreas (14%), adrenal gland (8%), CNS (8%), and bone (6%).
At a median follow-up of 37.3 months, 79.2% of patients were still alive and, at a median surveillance duration of 16.7 months, 71% of patients had begun a targeted therapy. There were a total of 17 deaths (33%).
Median progression-free survival was 16.6 months and median postsurveillance overall survival was 39.1 months.
During active surveillance, only four patients transitioned from good to intermediate IMDC prognostic class. IMDC classes overall maintained their prognostic value during active surveillance, Dr. Bimbatti said. IMDC class was also the only factor that was associated with time on surveillance.
At baseline, tumor burden was in one site for 65% of patients, two sites in 31%, and three or more sites for 4%, but during active surveillance, changes occurred in one site in 35% of patients, two in 48%, and more than two in 17%.
A change in tumor burden (greater than or equal to 2.2 times the original burden), however, was related to poorer postsurveillance survival (hazard ratio,1.23; P less than .01), but not with overall survival (HR, 1.0; P less than .05).
Conversely, any increase in metastatic sites was associated with both significantly worse postsurveillance and overall survival (HR = 2.6, P = 0.04; and HR = 3.3, P less than 0.01).
Commenting on the study, Primo N. Lara Jr., MD, of the University of California, Davis, Comprehensive Cancer Center explained that active surveillance “remains a reasonable option for highly selected patients.”
“[The investigators] provide reassuring data that active surveillance does not alter the IMDC risk grouping,” he said, and that the results of this study overall, were similar to those of prior research.
There were some limitations to the study, in that it was retrospective and conducted at a single institution. “It also failed to account for psychosocial issues such as anxiety during the active surveillance phase,” he said.
Critical questions also remain, Dr. Lara added, such as what are the validated selection criteria for patients entering active surveillance or what is the threshold of tumor burden to warrant active surveillance discontinuation.
The funding source was not disclosed. Dr. Bimbatti and his coauthors have no disclosures. Dr. Lara reports financial ties to multiple pharmaceutical companies.
ORLANDO – Active surveillance prior to initiating targeted therapy could be an option for some patients with metastatic renal cell carcinoma (mRCC), according to new findings.
“Active surveillance does not affect the efficacy of subsequent therapies,” said lead study author Davide Bimbatti, MD, of the Azienda Ospedaliera Universitaria Integrata, University of Verona (Italy). “In selected patients, active surveillance allows us to delay the start of systemic treatment, and patients in active surveillance rarely have a worsening of prognostic class,” he said in a press briefing held at the 2017 Genitourinary Cancers Symposium sponsored by the American Society of Clinical Oncology, ASTRO, and the Society of Urologic Oncology.
Targeted therapies can improve survival in mRCC but treatment is not curative and associated toxicities can interfere with quality of life and force patients to discontinue treatment. Active surveillance delays the use of systemic therapy and associated toxicities, and is a feasible strategy for patients with indolent disease.
However, the effect of active surveillance on tumor burden and prognosis have not been investigated.
To address this issue, Dr. Bimbatti and his colleagues conducted a retrospective study and evaluated the effect of active surveillance on overall survival and postsurveillance overall survival, progression-free survival, and tumor burden (a measure of the number of sites and tumor size).
The study cohort included 48 patients with mRCC who underwent active surveillance during 2007-2016 and changes in the International mRCC Database Consortium (IMDC) prognostic class and tumor burden were analyzed for associations with these endpoints.
At baseline, 69% of patients had a favorable prognostic IMDC class, 25% were at an intermediate class, and 6% had a poor class designation. The main sites of metastases were lung (56%), lymph nodes (25%), pancreas (14%), adrenal gland (8%), CNS (8%), and bone (6%).
At a median follow-up of 37.3 months, 79.2% of patients were still alive and, at a median surveillance duration of 16.7 months, 71% of patients had begun a targeted therapy. There were a total of 17 deaths (33%).
Median progression-free survival was 16.6 months and median postsurveillance overall survival was 39.1 months.
During active surveillance, only four patients transitioned from good to intermediate IMDC prognostic class. IMDC classes overall maintained their prognostic value during active surveillance, Dr. Bimbatti said. IMDC class was also the only factor that was associated with time on surveillance.
At baseline, tumor burden was in one site for 65% of patients, two sites in 31%, and three or more sites for 4%, but during active surveillance, changes occurred in one site in 35% of patients, two in 48%, and more than two in 17%.
A change in tumor burden (greater than or equal to 2.2 times the original burden), however, was related to poorer postsurveillance survival (hazard ratio,1.23; P less than .01), but not with overall survival (HR, 1.0; P less than .05).
Conversely, any increase in metastatic sites was associated with both significantly worse postsurveillance and overall survival (HR = 2.6, P = 0.04; and HR = 3.3, P less than 0.01).
Commenting on the study, Primo N. Lara Jr., MD, of the University of California, Davis, Comprehensive Cancer Center explained that active surveillance “remains a reasonable option for highly selected patients.”
“[The investigators] provide reassuring data that active surveillance does not alter the IMDC risk grouping,” he said, and that the results of this study overall, were similar to those of prior research.
There were some limitations to the study, in that it was retrospective and conducted at a single institution. “It also failed to account for psychosocial issues such as anxiety during the active surveillance phase,” he said.
Critical questions also remain, Dr. Lara added, such as what are the validated selection criteria for patients entering active surveillance or what is the threshold of tumor burden to warrant active surveillance discontinuation.
The funding source was not disclosed. Dr. Bimbatti and his coauthors have no disclosures. Dr. Lara reports financial ties to multiple pharmaceutical companies.
AT THE GENITOURINARY CANCERS SYMPOSIUM
Key clinical point: Active surveillance may be an option for some patients with metastatic renal cell carcinoma.
Major finding: A change in tumor burden (greater than or equal to 2.2 times the original burden), however, was related to poorer postsurveillance survival (HR,1.23; P less than .01) but not with overall survival (HR, 1.0; P less than.05).
Data source: Retrospective single center study that evaluated active surveillance in 48 patients with metastatic renal cell carcinoma.
Disclosures: The funding source was not disclosed. Dr. Bimbatti and his coauthors have no disclosures. Dr. Lara reports financial ties to multiple pharmaceutical companies.
Chemoradiation standard of care in muscle-invasive bladder cancer
Orlando – The updated results of a large phase III trial support the use of chemoradiation with 5-fluorouracil (5-FU) and mitomycin C (MMC) and confirm that this treatment regimen should be a standard of care for muscle-invasive bladder cancer (MIBC).
When comparing patients who received radiation therapy with those who received chemoradiation, there was a robust improvement in bladder cancer specific survival for the latter when adjusted for known prognostic factors (hazard ratio, 0.73; P = .043).
There was also a borderline significant improvement in metastasis-free survival (HR, 0.78) and a significant reduction in the need for salvage cystectomy in the patients treated with chemoradiation (2-year rate, chemoradiotherapy11% vs. radiation therapy:17%, HR, 0.54; P = .03).
There were no statistically significant differences between groups when it came to overall survival, but, even though overall survival did not reach significance, at 2 years, there was a hint of separation of the curves, explained study author Emma Hall, MD, from the Institute of Cancer Research, London at the 2017 Genitourinary Cancers Symposium sponsored by the American Society of Clinical Oncology, American Society for Radiation Oncology, and the Society of Urologic Oncology.
One of the treatment arms received a reduced rate of radiation therapy to see if that would decrease toxicity. “The radiation therapy volume modification that we used did not reduce toxicity, but there is no evidence of an increase in local failure rate, suggesting it is safe to pursue clinical trials of volume sparing radiation therapy using newer technology adaptive delivery techniques,” said Dr. Hall.
The initial findings of the BC2001 study showed that adding chemotherapy (5-FU + MMC) to radiotherapy significantly improved rates of MIBC locoregional control but that reduced high-dose volume versus standard radiotherapy did not significantly reduce late side effects.
This study was a clinical trial set up to test two different questions in the treatment of MIBC, as an alternative to cystectomy. “We wanted to see if adding synchronous chemotherapy to radiotherapy would improve locoregional recurrence control and if reducing the radiation dose to uninvolved bladder would reduce toxicity and not impact local regional recurrence control,” according to Dr. Hall.
Under the 2 x 2 partial factorial design, 458 patients were randomized to radiation therapy (n = 178) or chemoradiation (n = 182) and/or to standard radiation therapy (n = 108) or reduced high-dose volume radiation therapy (n = 111).
The primary endpoint was locoregional control, and secondary endpoints included overall survival, bladder-cancer specific survival, metastasis-free survival, and salvage cystectomy rates.
The initial patients received radiation therapy instead of chemoradiation, and there was a robust improvement in bladder cancer–specific survival when adjusted for known prognostic factors (HR, 0.73; P = .043).
The analysis, presented in 2012, showed a reduction of about one-third of locoregional recurrence. The local control rates were 54% in the radiotherapy-alone arm and 67% in the chemoradiotherapy arm.
There was no significant difference in overall survival at that time.
For the radiotherapy comparison, the rate of late toxicity was low, and much lower than was anticipated, at the outset of the trial, and there was no difference in treatment groups, said Dr. Hall.
In an updated analysis, with a median of 10 years of follow-up, 70% of the patients were now deceased. “These represent robust data, and it is unlikely we will see any changes to the data,” she noted.
The findings presented now had an additional 4 years of follow-up, and while there were additional late events, the results were basically the same.
The rate of local control now showed a 40% reduction in the risk of recurrence and 5-year local control rates of 49% in the radiotherapy arm and 63% in the chemoradiotherapy arm.
“With 10 years follow up, an improvement in locoregional control and a reduced salvage cystectomy rate is confirmed with chemoradiotherapy,” Dr. Hall concluded, “and, taken together with the good quality of life data we have, this is important for this group.”
In a discussion of the paper, Dr. Jonathan Rosenberg, MD, from Memorial Sloan Kettering Cancer Center in New York, agrees with the conclusion that the data continue to support the use of chemoradiotherapy and that 5-FU + MMC is a good option.
He noted that 5-FU + MMC is a standard of care regardless of cisplatin eligibility, but he cannot draw conclusions on dose volume. “There are also other options for chemosensitization,” he said, but it is also import to determine the best way to select patients who will derive the most benefit from chemoradiation.
“There is a high need for robust predictive biomarkers, and we need novel approaches to move beyond chemotherapy,” he said.
The study was supported by Cancer Research UK. Dr Hall has received research funding from Accuray, AstraZeneca, Aventis, and Bayer. Several co-authors also have disclosed relationships with industry. Dr. Rosenberg has disclosed multiple relationships with industry.
Orlando – The updated results of a large phase III trial support the use of chemoradiation with 5-fluorouracil (5-FU) and mitomycin C (MMC) and confirm that this treatment regimen should be a standard of care for muscle-invasive bladder cancer (MIBC).
When comparing patients who received radiation therapy with those who received chemoradiation, there was a robust improvement in bladder cancer specific survival for the latter when adjusted for known prognostic factors (hazard ratio, 0.73; P = .043).
There was also a borderline significant improvement in metastasis-free survival (HR, 0.78) and a significant reduction in the need for salvage cystectomy in the patients treated with chemoradiation (2-year rate, chemoradiotherapy11% vs. radiation therapy:17%, HR, 0.54; P = .03).
There were no statistically significant differences between groups when it came to overall survival, but, even though overall survival did not reach significance, at 2 years, there was a hint of separation of the curves, explained study author Emma Hall, MD, from the Institute of Cancer Research, London at the 2017 Genitourinary Cancers Symposium sponsored by the American Society of Clinical Oncology, American Society for Radiation Oncology, and the Society of Urologic Oncology.
One of the treatment arms received a reduced rate of radiation therapy to see if that would decrease toxicity. “The radiation therapy volume modification that we used did not reduce toxicity, but there is no evidence of an increase in local failure rate, suggesting it is safe to pursue clinical trials of volume sparing radiation therapy using newer technology adaptive delivery techniques,” said Dr. Hall.
The initial findings of the BC2001 study showed that adding chemotherapy (5-FU + MMC) to radiotherapy significantly improved rates of MIBC locoregional control but that reduced high-dose volume versus standard radiotherapy did not significantly reduce late side effects.
This study was a clinical trial set up to test two different questions in the treatment of MIBC, as an alternative to cystectomy. “We wanted to see if adding synchronous chemotherapy to radiotherapy would improve locoregional recurrence control and if reducing the radiation dose to uninvolved bladder would reduce toxicity and not impact local regional recurrence control,” according to Dr. Hall.
Under the 2 x 2 partial factorial design, 458 patients were randomized to radiation therapy (n = 178) or chemoradiation (n = 182) and/or to standard radiation therapy (n = 108) or reduced high-dose volume radiation therapy (n = 111).
The primary endpoint was locoregional control, and secondary endpoints included overall survival, bladder-cancer specific survival, metastasis-free survival, and salvage cystectomy rates.
The initial patients received radiation therapy instead of chemoradiation, and there was a robust improvement in bladder cancer–specific survival when adjusted for known prognostic factors (HR, 0.73; P = .043).
The analysis, presented in 2012, showed a reduction of about one-third of locoregional recurrence. The local control rates were 54% in the radiotherapy-alone arm and 67% in the chemoradiotherapy arm.
There was no significant difference in overall survival at that time.
For the radiotherapy comparison, the rate of late toxicity was low, and much lower than was anticipated, at the outset of the trial, and there was no difference in treatment groups, said Dr. Hall.
In an updated analysis, with a median of 10 years of follow-up, 70% of the patients were now deceased. “These represent robust data, and it is unlikely we will see any changes to the data,” she noted.
The findings presented now had an additional 4 years of follow-up, and while there were additional late events, the results were basically the same.
The rate of local control now showed a 40% reduction in the risk of recurrence and 5-year local control rates of 49% in the radiotherapy arm and 63% in the chemoradiotherapy arm.
“With 10 years follow up, an improvement in locoregional control and a reduced salvage cystectomy rate is confirmed with chemoradiotherapy,” Dr. Hall concluded, “and, taken together with the good quality of life data we have, this is important for this group.”
In a discussion of the paper, Dr. Jonathan Rosenberg, MD, from Memorial Sloan Kettering Cancer Center in New York, agrees with the conclusion that the data continue to support the use of chemoradiotherapy and that 5-FU + MMC is a good option.
He noted that 5-FU + MMC is a standard of care regardless of cisplatin eligibility, but he cannot draw conclusions on dose volume. “There are also other options for chemosensitization,” he said, but it is also import to determine the best way to select patients who will derive the most benefit from chemoradiation.
“There is a high need for robust predictive biomarkers, and we need novel approaches to move beyond chemotherapy,” he said.
The study was supported by Cancer Research UK. Dr Hall has received research funding from Accuray, AstraZeneca, Aventis, and Bayer. Several co-authors also have disclosed relationships with industry. Dr. Rosenberg has disclosed multiple relationships with industry.
Orlando – The updated results of a large phase III trial support the use of chemoradiation with 5-fluorouracil (5-FU) and mitomycin C (MMC) and confirm that this treatment regimen should be a standard of care for muscle-invasive bladder cancer (MIBC).
When comparing patients who received radiation therapy with those who received chemoradiation, there was a robust improvement in bladder cancer specific survival for the latter when adjusted for known prognostic factors (hazard ratio, 0.73; P = .043).
There was also a borderline significant improvement in metastasis-free survival (HR, 0.78) and a significant reduction in the need for salvage cystectomy in the patients treated with chemoradiation (2-year rate, chemoradiotherapy11% vs. radiation therapy:17%, HR, 0.54; P = .03).
There were no statistically significant differences between groups when it came to overall survival, but, even though overall survival did not reach significance, at 2 years, there was a hint of separation of the curves, explained study author Emma Hall, MD, from the Institute of Cancer Research, London at the 2017 Genitourinary Cancers Symposium sponsored by the American Society of Clinical Oncology, American Society for Radiation Oncology, and the Society of Urologic Oncology.
One of the treatment arms received a reduced rate of radiation therapy to see if that would decrease toxicity. “The radiation therapy volume modification that we used did not reduce toxicity, but there is no evidence of an increase in local failure rate, suggesting it is safe to pursue clinical trials of volume sparing radiation therapy using newer technology adaptive delivery techniques,” said Dr. Hall.
The initial findings of the BC2001 study showed that adding chemotherapy (5-FU + MMC) to radiotherapy significantly improved rates of MIBC locoregional control but that reduced high-dose volume versus standard radiotherapy did not significantly reduce late side effects.
This study was a clinical trial set up to test two different questions in the treatment of MIBC, as an alternative to cystectomy. “We wanted to see if adding synchronous chemotherapy to radiotherapy would improve locoregional recurrence control and if reducing the radiation dose to uninvolved bladder would reduce toxicity and not impact local regional recurrence control,” according to Dr. Hall.
Under the 2 x 2 partial factorial design, 458 patients were randomized to radiation therapy (n = 178) or chemoradiation (n = 182) and/or to standard radiation therapy (n = 108) or reduced high-dose volume radiation therapy (n = 111).
The primary endpoint was locoregional control, and secondary endpoints included overall survival, bladder-cancer specific survival, metastasis-free survival, and salvage cystectomy rates.
The initial patients received radiation therapy instead of chemoradiation, and there was a robust improvement in bladder cancer–specific survival when adjusted for known prognostic factors (HR, 0.73; P = .043).
The analysis, presented in 2012, showed a reduction of about one-third of locoregional recurrence. The local control rates were 54% in the radiotherapy-alone arm and 67% in the chemoradiotherapy arm.
There was no significant difference in overall survival at that time.
For the radiotherapy comparison, the rate of late toxicity was low, and much lower than was anticipated, at the outset of the trial, and there was no difference in treatment groups, said Dr. Hall.
In an updated analysis, with a median of 10 years of follow-up, 70% of the patients were now deceased. “These represent robust data, and it is unlikely we will see any changes to the data,” she noted.
The findings presented now had an additional 4 years of follow-up, and while there were additional late events, the results were basically the same.
The rate of local control now showed a 40% reduction in the risk of recurrence and 5-year local control rates of 49% in the radiotherapy arm and 63% in the chemoradiotherapy arm.
“With 10 years follow up, an improvement in locoregional control and a reduced salvage cystectomy rate is confirmed with chemoradiotherapy,” Dr. Hall concluded, “and, taken together with the good quality of life data we have, this is important for this group.”
In a discussion of the paper, Dr. Jonathan Rosenberg, MD, from Memorial Sloan Kettering Cancer Center in New York, agrees with the conclusion that the data continue to support the use of chemoradiotherapy and that 5-FU + MMC is a good option.
He noted that 5-FU + MMC is a standard of care regardless of cisplatin eligibility, but he cannot draw conclusions on dose volume. “There are also other options for chemosensitization,” he said, but it is also import to determine the best way to select patients who will derive the most benefit from chemoradiation.
“There is a high need for robust predictive biomarkers, and we need novel approaches to move beyond chemotherapy,” he said.
The study was supported by Cancer Research UK. Dr Hall has received research funding from Accuray, AstraZeneca, Aventis, and Bayer. Several co-authors also have disclosed relationships with industry. Dr. Rosenberg has disclosed multiple relationships with industry.
AT THE GENITOURINARY CANCERS SYMPOSIUM
Key clinical point: Chemoradiation with 5-FU + MMC should be a standard of care in muscle-invasive bladder cancer.
Major finding: When comparing radiation therapy versus chemoradiation, there was a robust improvement in bladder cancer specific survival when adjusted for known prognostic factors (HR, 0.73; P = .043).
Data source: A long-term phase III randomized trial that included 458 patients with MIBC.
Disclosures: The study was supported by Cancer Research UK. Dr. Hall has received research funding from Accuray, AstraZeneca, Aventis , and Bayer. Several coauthors also have disclosed relationships with industry. Dr. Rosenberg has disclosed multiple relationships with industry.
New significantly mutated genes detected in prostate cancer
A new large study has identified 78 significantly mutated genes (SMGs) and has enlarged the genetic landscape of prostate cancer. In addition, 37 genes that were not previously reported as SMGs in prostate cancer and 23 that were not previously identified as recurrently altered in cancer, were identified.
“Through aggregation and uniform genomic analysis, we refined the map of somatic mutations in prostate cancer and identified cancer genes and pathways not previously associated with this disease,” said lead author Dr. Joshua Armenia, of the Memorial Sloan Kettering Cancer Center, New York.
“Our findings may inform patient stratification and translational investigation,” Dr. Armenia said in a press briefing held in 2017 genitourinary cancers symposium sponsored by the American Society of Clinical Oncology, ASTRO, and the Society of Urologic Oncology.
The mutational landscapes of primary and metastatic prostate cancer have been robustly analyzed in multiple whole exome sequencing (WES) studies.
But prostate cancer is a very heterogenous disease, explained Dr. Armenia. Aggregation and uniform genomic analysis of larger cohorts can reveal significantly mutated genes and pathways in the “long tail” (1%-5% of cases).
The power to detect genes is significantly proportional to the number of samples used, and the size of the cohort. “Our knowledge of prostate cancer genes is incomplete,” said Dr. Armenia.
He and his colleagues hypothesized that an aggregate, uniform analysis of all data generated to date would enable discovery of new significantly mutated genes and pathways not previously associated with prostate cancer and shed more light onto the genetic differences between primary and metastatic prostate cancer.
They analyzed 918 tumors, with a total of 583 primary tumors and 335 metastasis, with a mutational significance analysis that uses statistical and biological approaches to determine which genes and pathways are recurrently altered.
This approach led to the identification of 78 SMGs, and 37 of the genes are novel prostate cancer genes implicated in other tumor types; SPEN, SETD2, ARID1A, CUL3 ARID2, SMARCAD1, U2AF1.
“These genes have never been implicated in prostate cancer but are found in other tumor types,” said Dr. Armenia. “We stratified them by pathway, and we found 21% of the tumors show mutations in epigenetic regulators and chromatin remodelers.”
The investigators found that 14% of these tumors show alterations in the ubiquitin pathway in prostate cancer. The novel mutations in CUL3 (M299R hotspot) are mutually exclusive with SPOP mutations.
“Unexpectedly, we also found alterations in splicing pathway alterations in prostate cancer,” Dr. Armenia noted.
There were 38 altered tumors (4% of the 918 total) that had alterations in genes such as U2AF1 (0.5%) and SF3B1 (1%), GEMINS (0.8%), TCERG1 (1.3%), and PRPFB (1.3%).
The researchers found another “interesting” gene, SPEN, which is a novel prostate cancer gene. It is a hormone regulator gene that has been described in breast cancer, where it is associated with tamoxifen resistance. Truncating mutations were found in SPEN, which is a hormone inducible transcriptional repressor, in 2.8% of samples which is a rate similar to the frequency observed in breast tumors. SPEN in prostate cancer may have a similar role to the one it has in breast cancer.
“We performed enrichment analysis of genomic alterations in metastatic tumors to be able to identify markers of advanced disease,” he explained. “And what we found that TP53, AR, PTEN, FOXA1, APC, and BRCA2 alterations are enriched in metastatic samples.”
Alterations in epigenetic regulators (KMT2C, KMT2D) are significantly enriched in metastatic tumors while SPOP mutations and FOXP1/RYBP deletions are enriched in primary tumors.
Dr. Armenia and colleagues also identified a subclass of epigenetically mutated prostate cancer, representing 21% of prostate cancers and insignificantly enriched tumors lacking an ETS fusion.
“We discovered novel pathways in prostate cancer including SW1/SNF and splicing, as well as novel prostate cancer genes that include CUL3 and SPEN, and a set of genomic markers enriched in advanced disease,” he concluded.
A new large study has identified 78 significantly mutated genes (SMGs) and has enlarged the genetic landscape of prostate cancer. In addition, 37 genes that were not previously reported as SMGs in prostate cancer and 23 that were not previously identified as recurrently altered in cancer, were identified.
“Through aggregation and uniform genomic analysis, we refined the map of somatic mutations in prostate cancer and identified cancer genes and pathways not previously associated with this disease,” said lead author Dr. Joshua Armenia, of the Memorial Sloan Kettering Cancer Center, New York.
“Our findings may inform patient stratification and translational investigation,” Dr. Armenia said in a press briefing held in 2017 genitourinary cancers symposium sponsored by the American Society of Clinical Oncology, ASTRO, and the Society of Urologic Oncology.
The mutational landscapes of primary and metastatic prostate cancer have been robustly analyzed in multiple whole exome sequencing (WES) studies.
But prostate cancer is a very heterogenous disease, explained Dr. Armenia. Aggregation and uniform genomic analysis of larger cohorts can reveal significantly mutated genes and pathways in the “long tail” (1%-5% of cases).
The power to detect genes is significantly proportional to the number of samples used, and the size of the cohort. “Our knowledge of prostate cancer genes is incomplete,” said Dr. Armenia.
He and his colleagues hypothesized that an aggregate, uniform analysis of all data generated to date would enable discovery of new significantly mutated genes and pathways not previously associated with prostate cancer and shed more light onto the genetic differences between primary and metastatic prostate cancer.
They analyzed 918 tumors, with a total of 583 primary tumors and 335 metastasis, with a mutational significance analysis that uses statistical and biological approaches to determine which genes and pathways are recurrently altered.
This approach led to the identification of 78 SMGs, and 37 of the genes are novel prostate cancer genes implicated in other tumor types; SPEN, SETD2, ARID1A, CUL3 ARID2, SMARCAD1, U2AF1.
“These genes have never been implicated in prostate cancer but are found in other tumor types,” said Dr. Armenia. “We stratified them by pathway, and we found 21% of the tumors show mutations in epigenetic regulators and chromatin remodelers.”
The investigators found that 14% of these tumors show alterations in the ubiquitin pathway in prostate cancer. The novel mutations in CUL3 (M299R hotspot) are mutually exclusive with SPOP mutations.
“Unexpectedly, we also found alterations in splicing pathway alterations in prostate cancer,” Dr. Armenia noted.
There were 38 altered tumors (4% of the 918 total) that had alterations in genes such as U2AF1 (0.5%) and SF3B1 (1%), GEMINS (0.8%), TCERG1 (1.3%), and PRPFB (1.3%).
The researchers found another “interesting” gene, SPEN, which is a novel prostate cancer gene. It is a hormone regulator gene that has been described in breast cancer, where it is associated with tamoxifen resistance. Truncating mutations were found in SPEN, which is a hormone inducible transcriptional repressor, in 2.8% of samples which is a rate similar to the frequency observed in breast tumors. SPEN in prostate cancer may have a similar role to the one it has in breast cancer.
“We performed enrichment analysis of genomic alterations in metastatic tumors to be able to identify markers of advanced disease,” he explained. “And what we found that TP53, AR, PTEN, FOXA1, APC, and BRCA2 alterations are enriched in metastatic samples.”
Alterations in epigenetic regulators (KMT2C, KMT2D) are significantly enriched in metastatic tumors while SPOP mutations and FOXP1/RYBP deletions are enriched in primary tumors.
Dr. Armenia and colleagues also identified a subclass of epigenetically mutated prostate cancer, representing 21% of prostate cancers and insignificantly enriched tumors lacking an ETS fusion.
“We discovered novel pathways in prostate cancer including SW1/SNF and splicing, as well as novel prostate cancer genes that include CUL3 and SPEN, and a set of genomic markers enriched in advanced disease,” he concluded.
A new large study has identified 78 significantly mutated genes (SMGs) and has enlarged the genetic landscape of prostate cancer. In addition, 37 genes that were not previously reported as SMGs in prostate cancer and 23 that were not previously identified as recurrently altered in cancer, were identified.
“Through aggregation and uniform genomic analysis, we refined the map of somatic mutations in prostate cancer and identified cancer genes and pathways not previously associated with this disease,” said lead author Dr. Joshua Armenia, of the Memorial Sloan Kettering Cancer Center, New York.
“Our findings may inform patient stratification and translational investigation,” Dr. Armenia said in a press briefing held in 2017 genitourinary cancers symposium sponsored by the American Society of Clinical Oncology, ASTRO, and the Society of Urologic Oncology.
The mutational landscapes of primary and metastatic prostate cancer have been robustly analyzed in multiple whole exome sequencing (WES) studies.
But prostate cancer is a very heterogenous disease, explained Dr. Armenia. Aggregation and uniform genomic analysis of larger cohorts can reveal significantly mutated genes and pathways in the “long tail” (1%-5% of cases).
The power to detect genes is significantly proportional to the number of samples used, and the size of the cohort. “Our knowledge of prostate cancer genes is incomplete,” said Dr. Armenia.
He and his colleagues hypothesized that an aggregate, uniform analysis of all data generated to date would enable discovery of new significantly mutated genes and pathways not previously associated with prostate cancer and shed more light onto the genetic differences between primary and metastatic prostate cancer.
They analyzed 918 tumors, with a total of 583 primary tumors and 335 metastasis, with a mutational significance analysis that uses statistical and biological approaches to determine which genes and pathways are recurrently altered.
This approach led to the identification of 78 SMGs, and 37 of the genes are novel prostate cancer genes implicated in other tumor types; SPEN, SETD2, ARID1A, CUL3 ARID2, SMARCAD1, U2AF1.
“These genes have never been implicated in prostate cancer but are found in other tumor types,” said Dr. Armenia. “We stratified them by pathway, and we found 21% of the tumors show mutations in epigenetic regulators and chromatin remodelers.”
The investigators found that 14% of these tumors show alterations in the ubiquitin pathway in prostate cancer. The novel mutations in CUL3 (M299R hotspot) are mutually exclusive with SPOP mutations.
“Unexpectedly, we also found alterations in splicing pathway alterations in prostate cancer,” Dr. Armenia noted.
There were 38 altered tumors (4% of the 918 total) that had alterations in genes such as U2AF1 (0.5%) and SF3B1 (1%), GEMINS (0.8%), TCERG1 (1.3%), and PRPFB (1.3%).
The researchers found another “interesting” gene, SPEN, which is a novel prostate cancer gene. It is a hormone regulator gene that has been described in breast cancer, where it is associated with tamoxifen resistance. Truncating mutations were found in SPEN, which is a hormone inducible transcriptional repressor, in 2.8% of samples which is a rate similar to the frequency observed in breast tumors. SPEN in prostate cancer may have a similar role to the one it has in breast cancer.
“We performed enrichment analysis of genomic alterations in metastatic tumors to be able to identify markers of advanced disease,” he explained. “And what we found that TP53, AR, PTEN, FOXA1, APC, and BRCA2 alterations are enriched in metastatic samples.”
Alterations in epigenetic regulators (KMT2C, KMT2D) are significantly enriched in metastatic tumors while SPOP mutations and FOXP1/RYBP deletions are enriched in primary tumors.
Dr. Armenia and colleagues also identified a subclass of epigenetically mutated prostate cancer, representing 21% of prostate cancers and insignificantly enriched tumors lacking an ETS fusion.
“We discovered novel pathways in prostate cancer including SW1/SNF and splicing, as well as novel prostate cancer genes that include CUL3 and SPEN, and a set of genomic markers enriched in advanced disease,” he concluded.
Key clinical point: New findings on prostate cancer genetics may help inform patient stratification and translational research.
Major finding: A total of 78 significantly mutated genes were identified in prostate cancer along with 37 genes that were not previously reported as significantly mutated.
Data source: An experimental study that analyzed 918 tumors obtained from prostate cancer patients.
Disclosures: The study does not list a funding source. Dr. Armenia has no disclosures but several coauthors report relationships with industry.
Prostate cancer susceptibility loci identified
ORLANDO – , and captured an additional 6% of the familial relevant risk for prostate cancer.
In the Oncoarray stratified analysis, the researchers detected two single nucleotide polymorphisms (SNPs) that were associated with early-onset prostate cancer, defined as disease occurring in men younger than 55 years, and four that were associated with both aggressive and indolent disease.
“By the end of this year we will have a genetic test with 170 variants, and men at the top of 1% of risk group would have a 5.72% risk over the average man,” she said. “But if we incorporated in the rarer variants, which are associated with very nasty disease, could we then start to identify men who are at risk for aggressive disease, and if so, how should we intensively screen them or even treat them?”
Currently genome-wide association studies (GWAS) have identified over 100 prostate cancer susceptibility loci, which included 33% of the prostate cancer familial relative risk in Europeans.
“We already know how to stratify populations into risk groups,” said Dr. Eeles. “It is important for us to do the right tests to see if they are really going to help in terms of targeted screening.”
The heritability of prostate cancer is 57%, but currently there are no common aggressive-disease markers.
To identify further susceptibility variants, Dr. Eeles and her colleagues conducted a prostate cancer GWAS that was far larger than previous investigations. Their analysis included 46,939 prostate cancer cases and 27,910 controls of European ancestry who were genotyped using the 35,369 prostate cancer cases and 33,164 controls of European ancestry that were generated using other large-scale genotyping arrays.
Dr. Eeles explained that, because they want their data to be useful for future investigations and “you can’t type everything as just too expensive – there are millions of SNPs across the genome – we put in a GWAS backbone.”
What that means, she explained, is that, when these are typed, it can be inferred that the other genotypes are using mathematical modeling.
Their Oncoarray was a large sample consisting of various ethnic groups, but in this presentation Dr. Eeles focused on an analysis of 98.500 European samples. The number reached 145,000 because they combined it with previous results in a meta-analysis of samples derived from men of both European and Asian descent.
The identification of additional loci also will enable fine mapping efforts into the underlying biology of prostate cancer susceptibility, she added.
There was no funding source disclosed. Dr. Eeles has received honoraria from Janssen and Succinct Communications. Coauthor Peter Kraft reported a relationship with Merck, but none of the other investigators had any disclosures.
ORLANDO – , and captured an additional 6% of the familial relevant risk for prostate cancer.
In the Oncoarray stratified analysis, the researchers detected two single nucleotide polymorphisms (SNPs) that were associated with early-onset prostate cancer, defined as disease occurring in men younger than 55 years, and four that were associated with both aggressive and indolent disease.
“By the end of this year we will have a genetic test with 170 variants, and men at the top of 1% of risk group would have a 5.72% risk over the average man,” she said. “But if we incorporated in the rarer variants, which are associated with very nasty disease, could we then start to identify men who are at risk for aggressive disease, and if so, how should we intensively screen them or even treat them?”
Currently genome-wide association studies (GWAS) have identified over 100 prostate cancer susceptibility loci, which included 33% of the prostate cancer familial relative risk in Europeans.
“We already know how to stratify populations into risk groups,” said Dr. Eeles. “It is important for us to do the right tests to see if they are really going to help in terms of targeted screening.”
The heritability of prostate cancer is 57%, but currently there are no common aggressive-disease markers.
To identify further susceptibility variants, Dr. Eeles and her colleagues conducted a prostate cancer GWAS that was far larger than previous investigations. Their analysis included 46,939 prostate cancer cases and 27,910 controls of European ancestry who were genotyped using the 35,369 prostate cancer cases and 33,164 controls of European ancestry that were generated using other large-scale genotyping arrays.
Dr. Eeles explained that, because they want their data to be useful for future investigations and “you can’t type everything as just too expensive – there are millions of SNPs across the genome – we put in a GWAS backbone.”
What that means, she explained, is that, when these are typed, it can be inferred that the other genotypes are using mathematical modeling.
Their Oncoarray was a large sample consisting of various ethnic groups, but in this presentation Dr. Eeles focused on an analysis of 98.500 European samples. The number reached 145,000 because they combined it with previous results in a meta-analysis of samples derived from men of both European and Asian descent.
The identification of additional loci also will enable fine mapping efforts into the underlying biology of prostate cancer susceptibility, she added.
There was no funding source disclosed. Dr. Eeles has received honoraria from Janssen and Succinct Communications. Coauthor Peter Kraft reported a relationship with Merck, but none of the other investigators had any disclosures.
ORLANDO – , and captured an additional 6% of the familial relevant risk for prostate cancer.
In the Oncoarray stratified analysis, the researchers detected two single nucleotide polymorphisms (SNPs) that were associated with early-onset prostate cancer, defined as disease occurring in men younger than 55 years, and four that were associated with both aggressive and indolent disease.
“By the end of this year we will have a genetic test with 170 variants, and men at the top of 1% of risk group would have a 5.72% risk over the average man,” she said. “But if we incorporated in the rarer variants, which are associated with very nasty disease, could we then start to identify men who are at risk for aggressive disease, and if so, how should we intensively screen them or even treat them?”
Currently genome-wide association studies (GWAS) have identified over 100 prostate cancer susceptibility loci, which included 33% of the prostate cancer familial relative risk in Europeans.
“We already know how to stratify populations into risk groups,” said Dr. Eeles. “It is important for us to do the right tests to see if they are really going to help in terms of targeted screening.”
The heritability of prostate cancer is 57%, but currently there are no common aggressive-disease markers.
To identify further susceptibility variants, Dr. Eeles and her colleagues conducted a prostate cancer GWAS that was far larger than previous investigations. Their analysis included 46,939 prostate cancer cases and 27,910 controls of European ancestry who were genotyped using the 35,369 prostate cancer cases and 33,164 controls of European ancestry that were generated using other large-scale genotyping arrays.
Dr. Eeles explained that, because they want their data to be useful for future investigations and “you can’t type everything as just too expensive – there are millions of SNPs across the genome – we put in a GWAS backbone.”
What that means, she explained, is that, when these are typed, it can be inferred that the other genotypes are using mathematical modeling.
Their Oncoarray was a large sample consisting of various ethnic groups, but in this presentation Dr. Eeles focused on an analysis of 98.500 European samples. The number reached 145,000 because they combined it with previous results in a meta-analysis of samples derived from men of both European and Asian descent.
The identification of additional loci also will enable fine mapping efforts into the underlying biology of prostate cancer susceptibility, she added.
There was no funding source disclosed. Dr. Eeles has received honoraria from Janssen and Succinct Communications. Coauthor Peter Kraft reported a relationship with Merck, but none of the other investigators had any disclosures.
AT THE GENITOURINARY CANCERS SYMPOSIUM
Key clinical point: Findings demonstrate the utility of high-density arrays and large sample sizes for novel genetic discovery.
Major finding: A meta-analysis from more than 145,000 men identified 65 novel prostate cancer susceptibility loci.
Data source: A large genomic study conducted to identify prostate cancer susceptibility loci.
Disclosures: There was no funding source disclosed. Dr. Eeles has received honoraria from Janssen and Succinct Communications. Coauthor Peter Kraft reported a relationship with Merck, but none of the other investigators had any disclosures to report.
Palliative care improves quality of life after HCT
Inpatient palliative care can help maintain quality of life (QoL) in patients undergoing hematopoietic stem cell transplantation (HCT), based on the results of a randomized clinical trial to assess the effect of inpatient palliative care on patient and caregiver-reported outcomes while hospitalized for HCT and for 3 months after transplantation.
During the 2-week period following their transplants, patients who received inpatient palliative care experienced a 14.72-point decrease in QoL, compared with a 21.54-point decrease in QoL for those assigned to standard transplant care alone. The difference was statistically significant (JAMA. 2016;316[20]:2094-2103. doi:10.1001/jama.2016.16786).
In addition to the QoL results, Areej El-Jawahri, MD, of Massachusetts General Hospital in Boston, and coauthors, noted that “exploratory secondary outcomes also showed that patients in the palliative care group benefited, with less increase in their depression symptoms, lower anxiety symptoms, and less increase in symptom burden compared with those receiving standard transplant care.
“Thus, palliative care may help to lessen the decline in QoL experienced by patients during hospitalization for HCT, which has long been perceived as a natural aspect of the transplantation process.”
The study cohort comprised 160 adults with hematologic malignancies undergoing autologous/allogeneic HCT, and 94 caregivers.
A total of 81 patients were assigned to the intervention, and were seen by palliative care clinicians at least twice a week while they were hospitalized. The palliative care intervention focused on managing both physical and psychological symptoms, and those who were assigned to the standard care arm (n = 79) could also request to be seen by the palliative care team.
Quality of life was measured based on mean FACT-BMT score. In the palliative care group, the FACT-BMT score was 110.26 at hospitalization and 95.46 at 2 weeks after transplant (mean change, −14.72). For the standard care group, FACT-BMT score was 106.83 at hospitalization and 85.42 at 2 weeks after transplant (mean change, −21.54) The −6.82 difference between the group groups was statistically significant (95% CI, −13.48 to −0.16; P = .045).
When looking at secondary outcomes, those in the intervention group had lower mean depression scores at 2 weeks based on the HADS-D measure. For the intervention group, the mean baseline score was 3.95 and the mean week 2 score was 6.39. For the control group, the baseline score was 4.94 and the week 2 score was 8.86. The difference between the groups was 1.49 (95% CI, 0.20-2.78; P = .02). Depression scores remained lower in the intervention group at 3 months.
The intervention group also reported a decrease in anxiety symptoms, but the control group reported an increase in anxiety symptoms from baseline to week 2 on the HADS-A measure. The mean difference in score between the two groups was 1.92; (95% CI, 0.83-3.01; P less than .001). However, there was no significant difference between the two groups at 3 months after transplant.
The study was supported by the National Palliative Care Research Foundation and grant K24 CA 181253 from the National Cancer Institute. Dr. El-Jawahri reported no disclosures.
Inpatient palliative care can help maintain quality of life (QoL) in patients undergoing hematopoietic stem cell transplantation (HCT), based on the results of a randomized clinical trial to assess the effect of inpatient palliative care on patient and caregiver-reported outcomes while hospitalized for HCT and for 3 months after transplantation.
During the 2-week period following their transplants, patients who received inpatient palliative care experienced a 14.72-point decrease in QoL, compared with a 21.54-point decrease in QoL for those assigned to standard transplant care alone. The difference was statistically significant (JAMA. 2016;316[20]:2094-2103. doi:10.1001/jama.2016.16786).
In addition to the QoL results, Areej El-Jawahri, MD, of Massachusetts General Hospital in Boston, and coauthors, noted that “exploratory secondary outcomes also showed that patients in the palliative care group benefited, with less increase in their depression symptoms, lower anxiety symptoms, and less increase in symptom burden compared with those receiving standard transplant care.
“Thus, palliative care may help to lessen the decline in QoL experienced by patients during hospitalization for HCT, which has long been perceived as a natural aspect of the transplantation process.”
The study cohort comprised 160 adults with hematologic malignancies undergoing autologous/allogeneic HCT, and 94 caregivers.
A total of 81 patients were assigned to the intervention, and were seen by palliative care clinicians at least twice a week while they were hospitalized. The palliative care intervention focused on managing both physical and psychological symptoms, and those who were assigned to the standard care arm (n = 79) could also request to be seen by the palliative care team.
Quality of life was measured based on mean FACT-BMT score. In the palliative care group, the FACT-BMT score was 110.26 at hospitalization and 95.46 at 2 weeks after transplant (mean change, −14.72). For the standard care group, FACT-BMT score was 106.83 at hospitalization and 85.42 at 2 weeks after transplant (mean change, −21.54) The −6.82 difference between the group groups was statistically significant (95% CI, −13.48 to −0.16; P = .045).
When looking at secondary outcomes, those in the intervention group had lower mean depression scores at 2 weeks based on the HADS-D measure. For the intervention group, the mean baseline score was 3.95 and the mean week 2 score was 6.39. For the control group, the baseline score was 4.94 and the week 2 score was 8.86. The difference between the groups was 1.49 (95% CI, 0.20-2.78; P = .02). Depression scores remained lower in the intervention group at 3 months.
The intervention group also reported a decrease in anxiety symptoms, but the control group reported an increase in anxiety symptoms from baseline to week 2 on the HADS-A measure. The mean difference in score between the two groups was 1.92; (95% CI, 0.83-3.01; P less than .001). However, there was no significant difference between the two groups at 3 months after transplant.
The study was supported by the National Palliative Care Research Foundation and grant K24 CA 181253 from the National Cancer Institute. Dr. El-Jawahri reported no disclosures.
Inpatient palliative care can help maintain quality of life (QoL) in patients undergoing hematopoietic stem cell transplantation (HCT), based on the results of a randomized clinical trial to assess the effect of inpatient palliative care on patient and caregiver-reported outcomes while hospitalized for HCT and for 3 months after transplantation.
During the 2-week period following their transplants, patients who received inpatient palliative care experienced a 14.72-point decrease in QoL, compared with a 21.54-point decrease in QoL for those assigned to standard transplant care alone. The difference was statistically significant (JAMA. 2016;316[20]:2094-2103. doi:10.1001/jama.2016.16786).
In addition to the QoL results, Areej El-Jawahri, MD, of Massachusetts General Hospital in Boston, and coauthors, noted that “exploratory secondary outcomes also showed that patients in the palliative care group benefited, with less increase in their depression symptoms, lower anxiety symptoms, and less increase in symptom burden compared with those receiving standard transplant care.
“Thus, palliative care may help to lessen the decline in QoL experienced by patients during hospitalization for HCT, which has long been perceived as a natural aspect of the transplantation process.”
The study cohort comprised 160 adults with hematologic malignancies undergoing autologous/allogeneic HCT, and 94 caregivers.
A total of 81 patients were assigned to the intervention, and were seen by palliative care clinicians at least twice a week while they were hospitalized. The palliative care intervention focused on managing both physical and psychological symptoms, and those who were assigned to the standard care arm (n = 79) could also request to be seen by the palliative care team.
Quality of life was measured based on mean FACT-BMT score. In the palliative care group, the FACT-BMT score was 110.26 at hospitalization and 95.46 at 2 weeks after transplant (mean change, −14.72). For the standard care group, FACT-BMT score was 106.83 at hospitalization and 85.42 at 2 weeks after transplant (mean change, −21.54) The −6.82 difference between the group groups was statistically significant (95% CI, −13.48 to −0.16; P = .045).
When looking at secondary outcomes, those in the intervention group had lower mean depression scores at 2 weeks based on the HADS-D measure. For the intervention group, the mean baseline score was 3.95 and the mean week 2 score was 6.39. For the control group, the baseline score was 4.94 and the week 2 score was 8.86. The difference between the groups was 1.49 (95% CI, 0.20-2.78; P = .02). Depression scores remained lower in the intervention group at 3 months.
The intervention group also reported a decrease in anxiety symptoms, but the control group reported an increase in anxiety symptoms from baseline to week 2 on the HADS-A measure. The mean difference in score between the two groups was 1.92; (95% CI, 0.83-3.01; P less than .001). However, there was no significant difference between the two groups at 3 months after transplant.
The study was supported by the National Palliative Care Research Foundation and grant K24 CA 181253 from the National Cancer Institute. Dr. El-Jawahri reported no disclosures.
FROM JAMA
Key clinical point: Inpatient palliative care can help maintain quality of life in patients undergoing hematopoietic stem cell transplantation.
Major finding: During the 2-week period following their transplants, patients who received inpatient palliative care experienced a 14.72-point decrease in QoL, compared with a 21.54-point decrease in QoL for those assigned to standard transplant care alone.
Data source: Randomized clinical trial among 160 adults with hematologic malignancies who underwent HCT, and their caregivers (n = 94).
Disclosures: The study was supported by the National Palliative Care Research Foundation and grant K24 CA 181253 from the National Cancer Institute. Dr. El-Jawahri reported no disclosures.
Decitabine elicits favorable response in high-risk AML/MDS
Single-agent therapy with decitabine elicited favorable responses in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) who had cytogenetic abnormalities associated with an unfavorable risk profile, a study showed.
Even though the responses were not long lasting, the overall survival rates were similar to those of AML patients who had an intermediate-risk cytogenetic profile and who had received the same treatment regimen.
Adult AML patients with karyotypes associated with unfavorable risk and older patients with AML (aged 60 or older) generally have poor outcomes, with a median survival in the range of 1 year. Those with AML and TP53 mutations tend to be older (median age, 61-67 years), and nearly all patients have karyotypes associated with unfavorable risk. These patients have particularly dismal outcomes, with median survival in the range of 4-6 months, if they receive cytotoxic chemotherapy.
In their study, John S Welch, MD, of Washington University, St Louis, and his colleagues evaluated somatic mutations and their relationships to clinical responses in 84 adult patients with AML or MDS who received treatment with decitabine as monotherapy.
Decitabine was administered at a dose of 20 mg/m2 of body surface area per day for 10 consecutive days in monthly cycles. An extension cohort that included 32 additional patients also were treated with decitabine but in different protocols.
Of the entire cohort of 116 patients, 15 patients (13%) achieved a complete remission, and 38 patients had bone marrow blast clearance with less than 5% blasts (complete remission with incomplete count recovery or morphologic complete remission). The overall response rate was 46%. In addition, 9 patients (8%), achieved a partial response, stable disease was observed in 23 patients (20%), and progressive disease was seen in 19 patients (16%).
Clinical responses were strongly correlated with karyotypes associated with unfavorable risk and the presence of TP53 mutations. Bone marrow blast clearance (complete remission, complete remission with incomplete count recovery, or morphologic complete remission) occurred in 29 of 43 patients with karyotypes associated with unfavorable risk (67%), compared with 24 of 71 patients with karyotypes associated with intermediate or favorable risk (34%). The same pattern was observed in all patients with TP53 mutations (100%) versus 32 of 78 patients with wildtype TP53 mutations (41%).
“Additional studies will be required to determine whether these differences in survival are truly due to improved responses associated with decitabine or whether conventional chemotherapy with an anthracycline and cytarabine actually decreases the rate of survival among patients with unfavorable-risk cytogenetic profiles,” wrote Dr. Welch and his colleagues.
The study was supported by the Specialized Program of Research Excellence in AML of the National Cancer Institute and the Genomics of AML Program Project. Dr. Welch had no disclosures, and several of his coauthors reported relationships with industry.
The current study raises the important question of the definition of a response. Complete remission conventionally entails bone marrow with less than 5% blasts and normalization of blood counts; absent these, remission is considered incomplete. After various confounding factors are taken into account, a complete response with cytotoxic therapy is associated with longer remissions and longer survival than is complete remission with incomplete count recovery.
The authors of this paper considered a response to be blast clearance to less than 5%, but complete remission was seen in only 4 of the 21 patients with TP53 mutations who fulfilled this criterion. The mutant allele burden was also similar in patients who had a response, regardless of blood count recovery.
In contrast, measurable residual disease is considerably more frequent in patients with complete remission with incomplete count recovery than in patients with complete remission who have received cytotoxic therapy, indicating that more data are needed on subsequent clinical outcomes according to whether clearance of blasts is accompanied by count recovery in patients with AML and TP53 mutations who have received decitabine.
AML “targeted-therapy” trials typically involve one drug, and this policy is called into question by the diverse molecular architecture (and brief remissions) observed in this trial. The trial by Welch et al. points to inevitable, rational replacement of large trials in which homogeneous therapy is administered for a heterogeneous disease by smaller, subgroup-specific trials.
The article also suggests questions that are likely to complicate this future.
Dr. Elihu Estey is with the division of hematology, University of Washington Medical Center, and the clinical research division, Fred Hutchinson Cancer Research Center, Seattle. He had no disclosures. These remarks were taken from an editorial accompanying Dr. Welch’s paper (N Engl J Med. 2016;375:2023-36).
The current study raises the important question of the definition of a response. Complete remission conventionally entails bone marrow with less than 5% blasts and normalization of blood counts; absent these, remission is considered incomplete. After various confounding factors are taken into account, a complete response with cytotoxic therapy is associated with longer remissions and longer survival than is complete remission with incomplete count recovery.
The authors of this paper considered a response to be blast clearance to less than 5%, but complete remission was seen in only 4 of the 21 patients with TP53 mutations who fulfilled this criterion. The mutant allele burden was also similar in patients who had a response, regardless of blood count recovery.
In contrast, measurable residual disease is considerably more frequent in patients with complete remission with incomplete count recovery than in patients with complete remission who have received cytotoxic therapy, indicating that more data are needed on subsequent clinical outcomes according to whether clearance of blasts is accompanied by count recovery in patients with AML and TP53 mutations who have received decitabine.
AML “targeted-therapy” trials typically involve one drug, and this policy is called into question by the diverse molecular architecture (and brief remissions) observed in this trial. The trial by Welch et al. points to inevitable, rational replacement of large trials in which homogeneous therapy is administered for a heterogeneous disease by smaller, subgroup-specific trials.
The article also suggests questions that are likely to complicate this future.
Dr. Elihu Estey is with the division of hematology, University of Washington Medical Center, and the clinical research division, Fred Hutchinson Cancer Research Center, Seattle. He had no disclosures. These remarks were taken from an editorial accompanying Dr. Welch’s paper (N Engl J Med. 2016;375:2023-36).
The current study raises the important question of the definition of a response. Complete remission conventionally entails bone marrow with less than 5% blasts and normalization of blood counts; absent these, remission is considered incomplete. After various confounding factors are taken into account, a complete response with cytotoxic therapy is associated with longer remissions and longer survival than is complete remission with incomplete count recovery.
The authors of this paper considered a response to be blast clearance to less than 5%, but complete remission was seen in only 4 of the 21 patients with TP53 mutations who fulfilled this criterion. The mutant allele burden was also similar in patients who had a response, regardless of blood count recovery.
In contrast, measurable residual disease is considerably more frequent in patients with complete remission with incomplete count recovery than in patients with complete remission who have received cytotoxic therapy, indicating that more data are needed on subsequent clinical outcomes according to whether clearance of blasts is accompanied by count recovery in patients with AML and TP53 mutations who have received decitabine.
AML “targeted-therapy” trials typically involve one drug, and this policy is called into question by the diverse molecular architecture (and brief remissions) observed in this trial. The trial by Welch et al. points to inevitable, rational replacement of large trials in which homogeneous therapy is administered for a heterogeneous disease by smaller, subgroup-specific trials.
The article also suggests questions that are likely to complicate this future.
Dr. Elihu Estey is with the division of hematology, University of Washington Medical Center, and the clinical research division, Fred Hutchinson Cancer Research Center, Seattle. He had no disclosures. These remarks were taken from an editorial accompanying Dr. Welch’s paper (N Engl J Med. 2016;375:2023-36).
Single-agent therapy with decitabine elicited favorable responses in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) who had cytogenetic abnormalities associated with an unfavorable risk profile, a study showed.
Even though the responses were not long lasting, the overall survival rates were similar to those of AML patients who had an intermediate-risk cytogenetic profile and who had received the same treatment regimen.
Adult AML patients with karyotypes associated with unfavorable risk and older patients with AML (aged 60 or older) generally have poor outcomes, with a median survival in the range of 1 year. Those with AML and TP53 mutations tend to be older (median age, 61-67 years), and nearly all patients have karyotypes associated with unfavorable risk. These patients have particularly dismal outcomes, with median survival in the range of 4-6 months, if they receive cytotoxic chemotherapy.
In their study, John S Welch, MD, of Washington University, St Louis, and his colleagues evaluated somatic mutations and their relationships to clinical responses in 84 adult patients with AML or MDS who received treatment with decitabine as monotherapy.
Decitabine was administered at a dose of 20 mg/m2 of body surface area per day for 10 consecutive days in monthly cycles. An extension cohort that included 32 additional patients also were treated with decitabine but in different protocols.
Of the entire cohort of 116 patients, 15 patients (13%) achieved a complete remission, and 38 patients had bone marrow blast clearance with less than 5% blasts (complete remission with incomplete count recovery or morphologic complete remission). The overall response rate was 46%. In addition, 9 patients (8%), achieved a partial response, stable disease was observed in 23 patients (20%), and progressive disease was seen in 19 patients (16%).
Clinical responses were strongly correlated with karyotypes associated with unfavorable risk and the presence of TP53 mutations. Bone marrow blast clearance (complete remission, complete remission with incomplete count recovery, or morphologic complete remission) occurred in 29 of 43 patients with karyotypes associated with unfavorable risk (67%), compared with 24 of 71 patients with karyotypes associated with intermediate or favorable risk (34%). The same pattern was observed in all patients with TP53 mutations (100%) versus 32 of 78 patients with wildtype TP53 mutations (41%).
“Additional studies will be required to determine whether these differences in survival are truly due to improved responses associated with decitabine or whether conventional chemotherapy with an anthracycline and cytarabine actually decreases the rate of survival among patients with unfavorable-risk cytogenetic profiles,” wrote Dr. Welch and his colleagues.
The study was supported by the Specialized Program of Research Excellence in AML of the National Cancer Institute and the Genomics of AML Program Project. Dr. Welch had no disclosures, and several of his coauthors reported relationships with industry.
Single-agent therapy with decitabine elicited favorable responses in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) who had cytogenetic abnormalities associated with an unfavorable risk profile, a study showed.
Even though the responses were not long lasting, the overall survival rates were similar to those of AML patients who had an intermediate-risk cytogenetic profile and who had received the same treatment regimen.
Adult AML patients with karyotypes associated with unfavorable risk and older patients with AML (aged 60 or older) generally have poor outcomes, with a median survival in the range of 1 year. Those with AML and TP53 mutations tend to be older (median age, 61-67 years), and nearly all patients have karyotypes associated with unfavorable risk. These patients have particularly dismal outcomes, with median survival in the range of 4-6 months, if they receive cytotoxic chemotherapy.
In their study, John S Welch, MD, of Washington University, St Louis, and his colleagues evaluated somatic mutations and their relationships to clinical responses in 84 adult patients with AML or MDS who received treatment with decitabine as monotherapy.
Decitabine was administered at a dose of 20 mg/m2 of body surface area per day for 10 consecutive days in monthly cycles. An extension cohort that included 32 additional patients also were treated with decitabine but in different protocols.
Of the entire cohort of 116 patients, 15 patients (13%) achieved a complete remission, and 38 patients had bone marrow blast clearance with less than 5% blasts (complete remission with incomplete count recovery or morphologic complete remission). The overall response rate was 46%. In addition, 9 patients (8%), achieved a partial response, stable disease was observed in 23 patients (20%), and progressive disease was seen in 19 patients (16%).
Clinical responses were strongly correlated with karyotypes associated with unfavorable risk and the presence of TP53 mutations. Bone marrow blast clearance (complete remission, complete remission with incomplete count recovery, or morphologic complete remission) occurred in 29 of 43 patients with karyotypes associated with unfavorable risk (67%), compared with 24 of 71 patients with karyotypes associated with intermediate or favorable risk (34%). The same pattern was observed in all patients with TP53 mutations (100%) versus 32 of 78 patients with wildtype TP53 mutations (41%).
“Additional studies will be required to determine whether these differences in survival are truly due to improved responses associated with decitabine or whether conventional chemotherapy with an anthracycline and cytarabine actually decreases the rate of survival among patients with unfavorable-risk cytogenetic profiles,” wrote Dr. Welch and his colleagues.
The study was supported by the Specialized Program of Research Excellence in AML of the National Cancer Institute and the Genomics of AML Program Project. Dr. Welch had no disclosures, and several of his coauthors reported relationships with industry.
FROM THE NEW ENGLAND JOURNAL OF MEDICINE
Key clinical point:
Major finding: Of 116 patients, 53 (46%) experienced bone marrow blast clearance (less than 5% blasts), and response rates were higher among those with an unfavorable cytogenetic risk profile.
Data source: A prospective single-center clinical trial that evaluated single-agent decitabine in 84 adult patients with AML or MDS, with an extension cohort.
Disclosures: The study was supported by the Specialized Program of Research Excellence in AML of the National Cancer Institute and the Genomics of AML Program Project. Dr. Welch had no disclosures, and several of his coauthors reported relationships with industry.
Worse outcomes for double-hit lymphomas after ASCT
Patients with double-hit lymphomas (DHLs) and double-expressor lymphomas (DELs) have inferior outcomes after undergoing autologous stem cell transplantation (ASCT), according to a new study published in the Journal of Clinical Oncology.
The worst outcomes were observed in patients with concurrent DELs and DHLs, and this “supports the concept that the double-hit/double-expressor biology appears to render DLBCL resistant to and less likely to be cured by chemotherapy,” write Alex Herrera, MD, an oncologist at the City of Hope, Duarte, Calif., and his colleagues.
But that said, a significant proportion of patients with relapsed/refractory DEL did experience durable remissions following ASCT, particularly those with isolated DEL without DHL.
This suggests that “the presence of DEL alone should not be considered a contraindication to ASCT,” the authors wrote (J Clin Oncol. 2016 Oct. 24. doi: 10.1200/JCO.2016.68.2740).
DHLs and DELs are subtypes of diffuse large B-cell lymphoma (DLBCL), and while they are associated with poor outcomes after standard chemoimmunotherapy, data remain limited as to outcomes of patients with relapsed or refractory disease who undergo ASCT.
The retrospective multicenter study included 117 patients with chemotherapy-sensitive relapsed/refractory DLBCL who underwent ASCT and had archival tumor material available. DEL with MYC/BCL2 coexpression was observed in 52 patients (44%) while 15 patients expressed MYC-R (13%), of whom 12 (10%) had DHL.
The median follow-up time was 45 months for survivors, and the 4-year progression-free survival (PFS) and overall survival (OS) were 54% for the entire cohort.
The 4-year PFS and OS in patients with DHL was worse as compared to those without DHL; 28% vs. 57% (P = .013), and 25% vs. 66% (P less than .001), respectively.
Those with DHL had poorer PFS (28%) and OS (25%), compared with patients with DEL but not DHL (PFS, 53% and OS, 61%) as well as patients with neither DEL nor DHL (PFS, 60% and OS, 70%; three-way P value for PFS, P = .013; OS, P = .002).
Patients with concurrent DEL and DHL had the poorest outcome, with a 4-year PFS of 0%.
After researchers adjusted for clinical characteristics, the only factors that remained significantly associated with PFS were DEL (hazard ratio, 1.8; P = .035) and DHL (HR, 2.9; P = .009). Factors that were significantly associated with OS were DHL (HR, 3.4; P = .004) and remission status at ASCT (HR for partial response, 2.4; P = .007).
Overall, patients with DHL were less likely to achieve a complete response following salvage therapy, and those with DEL and patients with DHL had a shorter time to relapse after induction therapy.
“Although some patients with relapsed/refractory DHL had long-term remission after ASCT (isolated DHL without DEL), the low survival rate in this group argues that alternative transplantation strategies, including allogeneic hematopoietic stem cell transplantation or peri-ASCT relapse prevention strategies should be studied,” they concluded.
Recognizing that the majority of patients with double-hit or double-expressor lymphoma will relapse after R-CHOP, this study evaluates the efficacy of autologous stem cell transplantation as a salvage modality. This is a carefully conducted, albeit retrospective, analysis of patients with relapsed or refractory double-hit or double-expressor lymphoma undergoing autologous hematopoietic stem cell transplantation (auto-HCT) at two high-volume institutions.
Although it is perhaps not surprising that double-hit and double-expressor phenotypes confer inferior outcomes, it is worth examining these issues in some detail. The first issue is that the authors have defined categories that are not recognized by the World Health Organization, but are routinely seen in clinical practice.
For example, it might be assumed that all patients with double-hit lymphoma will have MYC/BCL2 protein expression and, therefore, also have double-expressor lymphoma, but some patients with double-hit lymphoma do not have protein expression of MYC/BCL2 and these patients may have better outcomes than patients whose tumors display both double-hit and double-expressor characteristics.
A second caveat to interpreting the results is that the study population does not reflect the true denominator of all patients with relapsed diffuse large B-cell lymphoma, because only chemotherapy-sensitive patients undergoing auto-HCT were included.
So, should patients with relapsed double-hit and double-expressor lymphoma be offered auto-HCT? What are the alternatives to auto-HCT? Unfortunately, there are no clear answers to these questions, although the surprisingly excellent outcomes for patients without either of these features (70% long-term survival) suggest that there is a group of patients for whom auto-HCT remains an effective and standard tool. For double-hit and double-expressor lymphoma, a clinical trial based on specific biologic changes in individual patients is the ideal but is far from reality at this point.
Overall, despite being a retrospective series with a high attrition rate based on tissue availability, the central review of pathology, uniform assessment of double-hit and double-expressor features, and mature follow-up of 45 months makes this a thought-provoking and timely paper.
Sonali M. Smith, MD, is from the University of Chicago, and has disclosed a consulting or advisory role with Genentech, Seattle Genetics, TG Therapeutics, Gilead Sciences, Immunogenix, Pharmacyclics, NanoString Technologies, Genmab, Juno Therapeutics, Abbvie, and Portola Pharmaceuticals. These remarks were taken from the editorial accompanying Dr. Herrara’s report (J Clin Oncol. 2016 Oct 17. doi: 10.1200/JCO.2016.70.0625).
Recognizing that the majority of patients with double-hit or double-expressor lymphoma will relapse after R-CHOP, this study evaluates the efficacy of autologous stem cell transplantation as a salvage modality. This is a carefully conducted, albeit retrospective, analysis of patients with relapsed or refractory double-hit or double-expressor lymphoma undergoing autologous hematopoietic stem cell transplantation (auto-HCT) at two high-volume institutions.
Although it is perhaps not surprising that double-hit and double-expressor phenotypes confer inferior outcomes, it is worth examining these issues in some detail. The first issue is that the authors have defined categories that are not recognized by the World Health Organization, but are routinely seen in clinical practice.
For example, it might be assumed that all patients with double-hit lymphoma will have MYC/BCL2 protein expression and, therefore, also have double-expressor lymphoma, but some patients with double-hit lymphoma do not have protein expression of MYC/BCL2 and these patients may have better outcomes than patients whose tumors display both double-hit and double-expressor characteristics.
A second caveat to interpreting the results is that the study population does not reflect the true denominator of all patients with relapsed diffuse large B-cell lymphoma, because only chemotherapy-sensitive patients undergoing auto-HCT were included.
So, should patients with relapsed double-hit and double-expressor lymphoma be offered auto-HCT? What are the alternatives to auto-HCT? Unfortunately, there are no clear answers to these questions, although the surprisingly excellent outcomes for patients without either of these features (70% long-term survival) suggest that there is a group of patients for whom auto-HCT remains an effective and standard tool. For double-hit and double-expressor lymphoma, a clinical trial based on specific biologic changes in individual patients is the ideal but is far from reality at this point.
Overall, despite being a retrospective series with a high attrition rate based on tissue availability, the central review of pathology, uniform assessment of double-hit and double-expressor features, and mature follow-up of 45 months makes this a thought-provoking and timely paper.
Sonali M. Smith, MD, is from the University of Chicago, and has disclosed a consulting or advisory role with Genentech, Seattle Genetics, TG Therapeutics, Gilead Sciences, Immunogenix, Pharmacyclics, NanoString Technologies, Genmab, Juno Therapeutics, Abbvie, and Portola Pharmaceuticals. These remarks were taken from the editorial accompanying Dr. Herrara’s report (J Clin Oncol. 2016 Oct 17. doi: 10.1200/JCO.2016.70.0625).
Recognizing that the majority of patients with double-hit or double-expressor lymphoma will relapse after R-CHOP, this study evaluates the efficacy of autologous stem cell transplantation as a salvage modality. This is a carefully conducted, albeit retrospective, analysis of patients with relapsed or refractory double-hit or double-expressor lymphoma undergoing autologous hematopoietic stem cell transplantation (auto-HCT) at two high-volume institutions.
Although it is perhaps not surprising that double-hit and double-expressor phenotypes confer inferior outcomes, it is worth examining these issues in some detail. The first issue is that the authors have defined categories that are not recognized by the World Health Organization, but are routinely seen in clinical practice.
For example, it might be assumed that all patients with double-hit lymphoma will have MYC/BCL2 protein expression and, therefore, also have double-expressor lymphoma, but some patients with double-hit lymphoma do not have protein expression of MYC/BCL2 and these patients may have better outcomes than patients whose tumors display both double-hit and double-expressor characteristics.
A second caveat to interpreting the results is that the study population does not reflect the true denominator of all patients with relapsed diffuse large B-cell lymphoma, because only chemotherapy-sensitive patients undergoing auto-HCT were included.
So, should patients with relapsed double-hit and double-expressor lymphoma be offered auto-HCT? What are the alternatives to auto-HCT? Unfortunately, there are no clear answers to these questions, although the surprisingly excellent outcomes for patients without either of these features (70% long-term survival) suggest that there is a group of patients for whom auto-HCT remains an effective and standard tool. For double-hit and double-expressor lymphoma, a clinical trial based on specific biologic changes in individual patients is the ideal but is far from reality at this point.
Overall, despite being a retrospective series with a high attrition rate based on tissue availability, the central review of pathology, uniform assessment of double-hit and double-expressor features, and mature follow-up of 45 months makes this a thought-provoking and timely paper.
Sonali M. Smith, MD, is from the University of Chicago, and has disclosed a consulting or advisory role with Genentech, Seattle Genetics, TG Therapeutics, Gilead Sciences, Immunogenix, Pharmacyclics, NanoString Technologies, Genmab, Juno Therapeutics, Abbvie, and Portola Pharmaceuticals. These remarks were taken from the editorial accompanying Dr. Herrara’s report (J Clin Oncol. 2016 Oct 17. doi: 10.1200/JCO.2016.70.0625).
Patients with double-hit lymphomas (DHLs) and double-expressor lymphomas (DELs) have inferior outcomes after undergoing autologous stem cell transplantation (ASCT), according to a new study published in the Journal of Clinical Oncology.
The worst outcomes were observed in patients with concurrent DELs and DHLs, and this “supports the concept that the double-hit/double-expressor biology appears to render DLBCL resistant to and less likely to be cured by chemotherapy,” write Alex Herrera, MD, an oncologist at the City of Hope, Duarte, Calif., and his colleagues.
But that said, a significant proportion of patients with relapsed/refractory DEL did experience durable remissions following ASCT, particularly those with isolated DEL without DHL.
This suggests that “the presence of DEL alone should not be considered a contraindication to ASCT,” the authors wrote (J Clin Oncol. 2016 Oct. 24. doi: 10.1200/JCO.2016.68.2740).
DHLs and DELs are subtypes of diffuse large B-cell lymphoma (DLBCL), and while they are associated with poor outcomes after standard chemoimmunotherapy, data remain limited as to outcomes of patients with relapsed or refractory disease who undergo ASCT.
The retrospective multicenter study included 117 patients with chemotherapy-sensitive relapsed/refractory DLBCL who underwent ASCT and had archival tumor material available. DEL with MYC/BCL2 coexpression was observed in 52 patients (44%) while 15 patients expressed MYC-R (13%), of whom 12 (10%) had DHL.
The median follow-up time was 45 months for survivors, and the 4-year progression-free survival (PFS) and overall survival (OS) were 54% for the entire cohort.
The 4-year PFS and OS in patients with DHL was worse as compared to those without DHL; 28% vs. 57% (P = .013), and 25% vs. 66% (P less than .001), respectively.
Those with DHL had poorer PFS (28%) and OS (25%), compared with patients with DEL but not DHL (PFS, 53% and OS, 61%) as well as patients with neither DEL nor DHL (PFS, 60% and OS, 70%; three-way P value for PFS, P = .013; OS, P = .002).
Patients with concurrent DEL and DHL had the poorest outcome, with a 4-year PFS of 0%.
After researchers adjusted for clinical characteristics, the only factors that remained significantly associated with PFS were DEL (hazard ratio, 1.8; P = .035) and DHL (HR, 2.9; P = .009). Factors that were significantly associated with OS were DHL (HR, 3.4; P = .004) and remission status at ASCT (HR for partial response, 2.4; P = .007).
Overall, patients with DHL were less likely to achieve a complete response following salvage therapy, and those with DEL and patients with DHL had a shorter time to relapse after induction therapy.
“Although some patients with relapsed/refractory DHL had long-term remission after ASCT (isolated DHL without DEL), the low survival rate in this group argues that alternative transplantation strategies, including allogeneic hematopoietic stem cell transplantation or peri-ASCT relapse prevention strategies should be studied,” they concluded.
Patients with double-hit lymphomas (DHLs) and double-expressor lymphomas (DELs) have inferior outcomes after undergoing autologous stem cell transplantation (ASCT), according to a new study published in the Journal of Clinical Oncology.
The worst outcomes were observed in patients with concurrent DELs and DHLs, and this “supports the concept that the double-hit/double-expressor biology appears to render DLBCL resistant to and less likely to be cured by chemotherapy,” write Alex Herrera, MD, an oncologist at the City of Hope, Duarte, Calif., and his colleagues.
But that said, a significant proportion of patients with relapsed/refractory DEL did experience durable remissions following ASCT, particularly those with isolated DEL without DHL.
This suggests that “the presence of DEL alone should not be considered a contraindication to ASCT,” the authors wrote (J Clin Oncol. 2016 Oct. 24. doi: 10.1200/JCO.2016.68.2740).
DHLs and DELs are subtypes of diffuse large B-cell lymphoma (DLBCL), and while they are associated with poor outcomes after standard chemoimmunotherapy, data remain limited as to outcomes of patients with relapsed or refractory disease who undergo ASCT.
The retrospective multicenter study included 117 patients with chemotherapy-sensitive relapsed/refractory DLBCL who underwent ASCT and had archival tumor material available. DEL with MYC/BCL2 coexpression was observed in 52 patients (44%) while 15 patients expressed MYC-R (13%), of whom 12 (10%) had DHL.
The median follow-up time was 45 months for survivors, and the 4-year progression-free survival (PFS) and overall survival (OS) were 54% for the entire cohort.
The 4-year PFS and OS in patients with DHL was worse as compared to those without DHL; 28% vs. 57% (P = .013), and 25% vs. 66% (P less than .001), respectively.
Those with DHL had poorer PFS (28%) and OS (25%), compared with patients with DEL but not DHL (PFS, 53% and OS, 61%) as well as patients with neither DEL nor DHL (PFS, 60% and OS, 70%; three-way P value for PFS, P = .013; OS, P = .002).
Patients with concurrent DEL and DHL had the poorest outcome, with a 4-year PFS of 0%.
After researchers adjusted for clinical characteristics, the only factors that remained significantly associated with PFS were DEL (hazard ratio, 1.8; P = .035) and DHL (HR, 2.9; P = .009). Factors that were significantly associated with OS were DHL (HR, 3.4; P = .004) and remission status at ASCT (HR for partial response, 2.4; P = .007).
Overall, patients with DHL were less likely to achieve a complete response following salvage therapy, and those with DEL and patients with DHL had a shorter time to relapse after induction therapy.
“Although some patients with relapsed/refractory DHL had long-term remission after ASCT (isolated DHL without DEL), the low survival rate in this group argues that alternative transplantation strategies, including allogeneic hematopoietic stem cell transplantation or peri-ASCT relapse prevention strategies should be studied,” they concluded.
FROM THE JOURNAL OF CLINICAL ONCOLOGY
Key clinical point:
Major finding: The 4-year progression-free survival in patients with DEL vs. non-DEL was 48% versus 59% (P = .049), and the 4-year OS was 56% vs. 67% (P = .10).
Data source: Retrospective, multicenter study that included 117 patients with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma who underwent ASCT.
Disclosures: The study was funded by a Conquer Cancer Foundation/ASCO Young Investigator Award and National Cancer Institute Grants; the Dana-Farber Cancer Institute Award Fund for Collaborative Research Initiatives in Hematologic Oncology; the Harold and Virginia Lash/David Lash Fund for Lymphoma Research; and NCI Grant No. P30CA033572 for work performed in the COH Pathology Core. Dr. Herrera reports receiving research funding from Seattle Genetics, Pharmacyclics, Genentech, Immune Design, and Sequenta, and received travel, accommodations, and expenses from Bristol-Myers Squibb. Several coauthors also report relationships with industry.
HCT survivors experience high rates of late respiratory and infectious complications
Cancer survivors who underwent hematopoietic cell transplantation (HCT) face a greater risk for hospitalizations and mortality, compared with survivors who did not have HCT.
New findings show that disparities in infectious and respiratory complications were marked between the two groups, but differences in circulatory disease, mental health diagnoses, and second cancers were insignificant.
“Clinicians who care for long-term survivors of HCT should be aware of comprehensive surveillance guidelines available for this high-risk population,” wrote Eric J. Chow, MD, of the Fred Hutchinson Cancer Research Center, Seattle, and his colleagues (J Clin Oncol. 2016 Nov 21. doi: 10.1200/JCO.2016.68.8457).
There have only been a few comprehensive analyses that have compared HCT with non-HCT cancer survivors. Thus, the authors noted that it is unclear if HCT survivors are at a greater risk of late complications, compared with other cancer survivors.
To address this issue, Dr. Chow and his team matched 2-year cancer survivors who had undergone HCT (n = 1,792; 52% allogeneic and 90% hematologic malignancies) to non-HCT 2-year cancer survivors, using a state cancer registry (n = 5,455), and the general population (n = 16,340), using driver’s license files.
The investigators found that the 10-year cumulative incidence of any hospitalization or death related to all major organ-system outcomes was significantly different (P less than .05) between the HCT survivors and general population.
Patients with a history of HCT had a 30.6% cumulative incidence of infectious complications (difference vs. non-HCT: 8.7%) and a 26.8% incidence of any respiratory complications (difference vs. non-HCT: 6.9%), the investigators reported.
In contrast, the 10-year cumulative incidences of nervous system, circulatory, and genitourinary complications; mental health outcomes; and the development of new cancers did not differ between the HCT and non-HCT groups.
The incidence of pregnancy-related hospitalization among women of childbearing age was lower in the HCT group, compared with non-HCT patients (group difference, 24.4%).
At the 2-year endpoint, Dr. Chow and his associates noted that certain risks were “notably higher” in patients who had undergone HCT, including primary infections (hazard ratio, 1.4), respiratory complications (HR, 1.3), and death from any cause (HR, 1.1).
A significantly greater hospitalization rate also was observed in the HCT group versus the non-HCT group (280 episodes per 1,000 person-years vs. 173 episodes per 1,000 person years; P less than .001).
“Future work to identify more specific risk factors associated with late infections and respiratory complications may help to further refine these guidelines and identify new prevention strategies,” the authors concluded.
The study was funded by grants from the National Institutes of Health. Dr. Chow had no disclosures and several coauthors report relationships with industry.
Cancer survivors who underwent hematopoietic cell transplantation (HCT) face a greater risk for hospitalizations and mortality, compared with survivors who did not have HCT.
New findings show that disparities in infectious and respiratory complications were marked between the two groups, but differences in circulatory disease, mental health diagnoses, and second cancers were insignificant.
“Clinicians who care for long-term survivors of HCT should be aware of comprehensive surveillance guidelines available for this high-risk population,” wrote Eric J. Chow, MD, of the Fred Hutchinson Cancer Research Center, Seattle, and his colleagues (J Clin Oncol. 2016 Nov 21. doi: 10.1200/JCO.2016.68.8457).
There have only been a few comprehensive analyses that have compared HCT with non-HCT cancer survivors. Thus, the authors noted that it is unclear if HCT survivors are at a greater risk of late complications, compared with other cancer survivors.
To address this issue, Dr. Chow and his team matched 2-year cancer survivors who had undergone HCT (n = 1,792; 52% allogeneic and 90% hematologic malignancies) to non-HCT 2-year cancer survivors, using a state cancer registry (n = 5,455), and the general population (n = 16,340), using driver’s license files.
The investigators found that the 10-year cumulative incidence of any hospitalization or death related to all major organ-system outcomes was significantly different (P less than .05) between the HCT survivors and general population.
Patients with a history of HCT had a 30.6% cumulative incidence of infectious complications (difference vs. non-HCT: 8.7%) and a 26.8% incidence of any respiratory complications (difference vs. non-HCT: 6.9%), the investigators reported.
In contrast, the 10-year cumulative incidences of nervous system, circulatory, and genitourinary complications; mental health outcomes; and the development of new cancers did not differ between the HCT and non-HCT groups.
The incidence of pregnancy-related hospitalization among women of childbearing age was lower in the HCT group, compared with non-HCT patients (group difference, 24.4%).
At the 2-year endpoint, Dr. Chow and his associates noted that certain risks were “notably higher” in patients who had undergone HCT, including primary infections (hazard ratio, 1.4), respiratory complications (HR, 1.3), and death from any cause (HR, 1.1).
A significantly greater hospitalization rate also was observed in the HCT group versus the non-HCT group (280 episodes per 1,000 person-years vs. 173 episodes per 1,000 person years; P less than .001).
“Future work to identify more specific risk factors associated with late infections and respiratory complications may help to further refine these guidelines and identify new prevention strategies,” the authors concluded.
The study was funded by grants from the National Institutes of Health. Dr. Chow had no disclosures and several coauthors report relationships with industry.
Cancer survivors who underwent hematopoietic cell transplantation (HCT) face a greater risk for hospitalizations and mortality, compared with survivors who did not have HCT.
New findings show that disparities in infectious and respiratory complications were marked between the two groups, but differences in circulatory disease, mental health diagnoses, and second cancers were insignificant.
“Clinicians who care for long-term survivors of HCT should be aware of comprehensive surveillance guidelines available for this high-risk population,” wrote Eric J. Chow, MD, of the Fred Hutchinson Cancer Research Center, Seattle, and his colleagues (J Clin Oncol. 2016 Nov 21. doi: 10.1200/JCO.2016.68.8457).
There have only been a few comprehensive analyses that have compared HCT with non-HCT cancer survivors. Thus, the authors noted that it is unclear if HCT survivors are at a greater risk of late complications, compared with other cancer survivors.
To address this issue, Dr. Chow and his team matched 2-year cancer survivors who had undergone HCT (n = 1,792; 52% allogeneic and 90% hematologic malignancies) to non-HCT 2-year cancer survivors, using a state cancer registry (n = 5,455), and the general population (n = 16,340), using driver’s license files.
The investigators found that the 10-year cumulative incidence of any hospitalization or death related to all major organ-system outcomes was significantly different (P less than .05) between the HCT survivors and general population.
Patients with a history of HCT had a 30.6% cumulative incidence of infectious complications (difference vs. non-HCT: 8.7%) and a 26.8% incidence of any respiratory complications (difference vs. non-HCT: 6.9%), the investigators reported.
In contrast, the 10-year cumulative incidences of nervous system, circulatory, and genitourinary complications; mental health outcomes; and the development of new cancers did not differ between the HCT and non-HCT groups.
The incidence of pregnancy-related hospitalization among women of childbearing age was lower in the HCT group, compared with non-HCT patients (group difference, 24.4%).
At the 2-year endpoint, Dr. Chow and his associates noted that certain risks were “notably higher” in patients who had undergone HCT, including primary infections (hazard ratio, 1.4), respiratory complications (HR, 1.3), and death from any cause (HR, 1.1).
A significantly greater hospitalization rate also was observed in the HCT group versus the non-HCT group (280 episodes per 1,000 person-years vs. 173 episodes per 1,000 person years; P less than .001).
“Future work to identify more specific risk factors associated with late infections and respiratory complications may help to further refine these guidelines and identify new prevention strategies,” the authors concluded.
The study was funded by grants from the National Institutes of Health. Dr. Chow had no disclosures and several coauthors report relationships with industry.
FROM THE JOURNAL OF CLINICAL ONCOLOGY
Key clinical point: Clinicians who care for HCT survivors should be aware of their high rates of late respiratory and infectious complications.
Major finding: Patients with a history of HCT had a 30.6% cumulative incidence of infectious complications (difference vs. non-HCT: 8.7%) and a 26.8% incidence of any respiratory complications (difference vs. non-HCT: 6.9%).
Data source: Retrospective population study using databases to match outcomes between two patient groups and the general population.
Disclosures: The study was funded by grants from the National Institutes of Health. Dr. Chow has no disclosures and several coauthors report relationships with industry.
ADT not associated with increased risk of dementia in larger study
Treatment with androgen deprivation therapy (ADT) was not associated with an increased risk of dementia in a large, population-based study, according to investigators.
Previous studies have demonstrated that low testosterone levels are associated with cognitive impairment and Alzheimer’s disease, but the association between ADT and cognitive impairment remains controversial, with conflicting results being reported.
In the current large population-based study, compared with nonuse, treatment with ADT did not increase the risk of dementia (incidence, 7.4 vs. 4.4 per 1,000 person years, adjusted hazard ratio, 1.02; 95%CI, 0.87-1.19), reported Farzin Khosrow-Khavar, PhD, of McGill University, Montreal, and colleagues.
“Consistent with our findings, a recent meta-analysis found that although patients treated with ADT performed worse on visuomotor tasks in comparison with control subjects or their own baseline assessments, there were no significant differences in cognitive domains that are pertinent to dementia including performance on attention/working memory, executive function, language, verbal memory, visual memory, and visuospatial ability” they wrote (J Clin Oncol. 2016 Nov. 21 doi: 10.1200/JCO.2016.69.6203).
In a smaller study of 9,272 patients, recently published in JAMA Oncology, prostate cancer patients treated with ADT were more than twice as likely as were those who were not to develop dementia.
For the current study, investigators identified a cohort of 30,903 men who were newly diagnosed with nonmetastatic prostate cancer between April 1, 1988, and April 30, 2015, using the United Kingdom’s Clinical Practice Research Datalink. The patients were then observed until April 30, 2016.
Within this cohort, 799 patients were newly diagnosed with dementia, which extrapolated to a crude incidence of 6.0 per 1,000 person-years. During the follow-up period, 17,994 patients (58.2%) were treated with ADT, and the median duration of use was 2.3 years.
Men who were using ADT tended to be older and were more likely to have ever used tobacco products, and they were also more likely to have had higher prostate-specific antigen levels and a higher prevalence of comorbidities, compared with nonusers.
In the primary analysis, ADT use was not associated with an overall higher risk of developing dementia (adjusted HR, 1.02; 95% CI, 0.87-1.19).
In secondary analyses, the risk did not vary with cumulative duration of use. Findings were similar when the risk was assessed by the type of ADT used for treatment, and when the association was evaluated with Alzheimer’s disease (adjusted HR, 1.11; 95% CI, 0.85-1.44) versus other types of dementia (adjusted HR, 0.97; 95% CI, 0.80-1.18).
The authors noted that additional studies in different settings are needed to confirm these new findings.
The study was supported by a foundation grant from the Canadian Institutes of Health Research. The authors declared that there were no conflicts of interest.
Treatment with androgen deprivation therapy (ADT) was not associated with an increased risk of dementia in a large, population-based study, according to investigators.
Previous studies have demonstrated that low testosterone levels are associated with cognitive impairment and Alzheimer’s disease, but the association between ADT and cognitive impairment remains controversial, with conflicting results being reported.
In the current large population-based study, compared with nonuse, treatment with ADT did not increase the risk of dementia (incidence, 7.4 vs. 4.4 per 1,000 person years, adjusted hazard ratio, 1.02; 95%CI, 0.87-1.19), reported Farzin Khosrow-Khavar, PhD, of McGill University, Montreal, and colleagues.
“Consistent with our findings, a recent meta-analysis found that although patients treated with ADT performed worse on visuomotor tasks in comparison with control subjects or their own baseline assessments, there were no significant differences in cognitive domains that are pertinent to dementia including performance on attention/working memory, executive function, language, verbal memory, visual memory, and visuospatial ability” they wrote (J Clin Oncol. 2016 Nov. 21 doi: 10.1200/JCO.2016.69.6203).
In a smaller study of 9,272 patients, recently published in JAMA Oncology, prostate cancer patients treated with ADT were more than twice as likely as were those who were not to develop dementia.
For the current study, investigators identified a cohort of 30,903 men who were newly diagnosed with nonmetastatic prostate cancer between April 1, 1988, and April 30, 2015, using the United Kingdom’s Clinical Practice Research Datalink. The patients were then observed until April 30, 2016.
Within this cohort, 799 patients were newly diagnosed with dementia, which extrapolated to a crude incidence of 6.0 per 1,000 person-years. During the follow-up period, 17,994 patients (58.2%) were treated with ADT, and the median duration of use was 2.3 years.
Men who were using ADT tended to be older and were more likely to have ever used tobacco products, and they were also more likely to have had higher prostate-specific antigen levels and a higher prevalence of comorbidities, compared with nonusers.
In the primary analysis, ADT use was not associated with an overall higher risk of developing dementia (adjusted HR, 1.02; 95% CI, 0.87-1.19).
In secondary analyses, the risk did not vary with cumulative duration of use. Findings were similar when the risk was assessed by the type of ADT used for treatment, and when the association was evaluated with Alzheimer’s disease (adjusted HR, 1.11; 95% CI, 0.85-1.44) versus other types of dementia (adjusted HR, 0.97; 95% CI, 0.80-1.18).
The authors noted that additional studies in different settings are needed to confirm these new findings.
The study was supported by a foundation grant from the Canadian Institutes of Health Research. The authors declared that there were no conflicts of interest.
Treatment with androgen deprivation therapy (ADT) was not associated with an increased risk of dementia in a large, population-based study, according to investigators.
Previous studies have demonstrated that low testosterone levels are associated with cognitive impairment and Alzheimer’s disease, but the association between ADT and cognitive impairment remains controversial, with conflicting results being reported.
In the current large population-based study, compared with nonuse, treatment with ADT did not increase the risk of dementia (incidence, 7.4 vs. 4.4 per 1,000 person years, adjusted hazard ratio, 1.02; 95%CI, 0.87-1.19), reported Farzin Khosrow-Khavar, PhD, of McGill University, Montreal, and colleagues.
“Consistent with our findings, a recent meta-analysis found that although patients treated with ADT performed worse on visuomotor tasks in comparison with control subjects or their own baseline assessments, there were no significant differences in cognitive domains that are pertinent to dementia including performance on attention/working memory, executive function, language, verbal memory, visual memory, and visuospatial ability” they wrote (J Clin Oncol. 2016 Nov. 21 doi: 10.1200/JCO.2016.69.6203).
In a smaller study of 9,272 patients, recently published in JAMA Oncology, prostate cancer patients treated with ADT were more than twice as likely as were those who were not to develop dementia.
For the current study, investigators identified a cohort of 30,903 men who were newly diagnosed with nonmetastatic prostate cancer between April 1, 1988, and April 30, 2015, using the United Kingdom’s Clinical Practice Research Datalink. The patients were then observed until April 30, 2016.
Within this cohort, 799 patients were newly diagnosed with dementia, which extrapolated to a crude incidence of 6.0 per 1,000 person-years. During the follow-up period, 17,994 patients (58.2%) were treated with ADT, and the median duration of use was 2.3 years.
Men who were using ADT tended to be older and were more likely to have ever used tobacco products, and they were also more likely to have had higher prostate-specific antigen levels and a higher prevalence of comorbidities, compared with nonusers.
In the primary analysis, ADT use was not associated with an overall higher risk of developing dementia (adjusted HR, 1.02; 95% CI, 0.87-1.19).
In secondary analyses, the risk did not vary with cumulative duration of use. Findings were similar when the risk was assessed by the type of ADT used for treatment, and when the association was evaluated with Alzheimer’s disease (adjusted HR, 1.11; 95% CI, 0.85-1.44) versus other types of dementia (adjusted HR, 0.97; 95% CI, 0.80-1.18).
The authors noted that additional studies in different settings are needed to confirm these new findings.
The study was supported by a foundation grant from the Canadian Institutes of Health Research. The authors declared that there were no conflicts of interest.
FROM JOURNAL OF CLINICAL ONCOLOGY
Key clinical point:
Major finding: Compared with nonuse of ADT, the use of ADT was not associated with an increased risk of dementia (incidence, 7.4 vs. 4.4 per 1,000 person-years, HR, 1.02; 95% CI, 0.87-1.19).
Data source: Population-based study that included 30,903 men newly diagnosed with nonmetastatic prostate cancer.
Disclosures: The study was supported by a foundation grant from the Canadian Institutes of Health Research. The authors declared that there were no conflicts of interest.
DNA methylation markers prognostic for metastatic breast cancer
A novel panel of DNA methylation markers may aid in predicting survival outcomes in metastatic breast cancer (MBC), according to new findings.
Using a new quantitative assay known as cMethDNA, researchers found that a high cumulative methylation index (CMI) level, as measured with a six-gene panel after beginning a new treatment, was consistently associated with both progression-free survival and overall survival, as well as progressive disease.
Even though these results are encouraging, however, its clinical application is still unknown, they noted.
“Although monitoring for the change in the CMI for treatment response has prognostic usefulness, its clinical usefulness in influencing changes in therapy must now be evaluated formally in randomized clinical trials,” wrote Kala Visvanathan, MD, of Johns Hopkins University, Baltimore, and her colleagues. “Furthermore, whether there is meaningful risk reclassification of patients with the addition of the CMI is worth careful assessment in future larger validation studies.”
Epigenetic alterations are common in human malignancies, and DNA methylation is a form of epigenetic alteration that is heritable during DNA replication. The authors developed a highly sensitive, high-throughput quantitative multiplex methylation-specific polymerase chain reaction assay known as cMethDNA that was able to detect circulating cell-free methylated DNA.
In this study, 10 genes were tested from 141 women with MBC who were beginning a new systemic therapy and who received treatment at participating academic medical centers.
Among patients with a high CMI, the median progression-free survival and overall survival were significantly shorter (PFS, 2.1 months; OS, 12.3 months), compared with those with a low CMI (PFS, 5.8 months; OS, 21.7 months), Dr. Visvanathan and her associates reported (J Clin Oncol. 2016 Nov. 21 doi: 10.1200/JCO.2015.66.2080).
Upon multivariable analysis, a high CMI versus low CMI at 4 weeks was independently associated with poorer PFS (hazard ratio, 1.79; 95% CI, 1.23-2.60; P = .002) and OS (hazard ratio, 1.75; 95% CI, 1.21-2.54; P = .003). Rising CMI levels from baseline to week 4 also were associated with worse PFS (P less than .001) as well as a 4.6-fold increase in the risk of progressive disease at first restaging (OR, 4.58; 95% CI, 1.82-11.60; P = .001) when compared with either a drop in CMI or no change at all.
The CMI level at week 4 was observed to be a strong predictor of PFS and improved the prediction of the base model, even in the presence of circulating tumor cells (P = .004).
Dr. Visvanathan and her colleagues concluded that these findings “must be validated to determine the clinical usefulness of the cMethDNA assay for specific treatments and tumor phenotypes in patients with metastatic disease and early-stage breast cancer.”
The study was supported by the Avon Foundation for Women, the Breast Cancer Research Foundation, Janssen Diagnostics, the Rubenstein Family Fund, the Susan G. Komen Foundation, and National Institutes of Health Grants. Dr. Visvanathan has declared holding patents, royalties, and other intellectual property, and several coauthors also have declared relationships with industry.
A novel panel of DNA methylation markers may aid in predicting survival outcomes in metastatic breast cancer (MBC), according to new findings.
Using a new quantitative assay known as cMethDNA, researchers found that a high cumulative methylation index (CMI) level, as measured with a six-gene panel after beginning a new treatment, was consistently associated with both progression-free survival and overall survival, as well as progressive disease.
Even though these results are encouraging, however, its clinical application is still unknown, they noted.
“Although monitoring for the change in the CMI for treatment response has prognostic usefulness, its clinical usefulness in influencing changes in therapy must now be evaluated formally in randomized clinical trials,” wrote Kala Visvanathan, MD, of Johns Hopkins University, Baltimore, and her colleagues. “Furthermore, whether there is meaningful risk reclassification of patients with the addition of the CMI is worth careful assessment in future larger validation studies.”
Epigenetic alterations are common in human malignancies, and DNA methylation is a form of epigenetic alteration that is heritable during DNA replication. The authors developed a highly sensitive, high-throughput quantitative multiplex methylation-specific polymerase chain reaction assay known as cMethDNA that was able to detect circulating cell-free methylated DNA.
In this study, 10 genes were tested from 141 women with MBC who were beginning a new systemic therapy and who received treatment at participating academic medical centers.
Among patients with a high CMI, the median progression-free survival and overall survival were significantly shorter (PFS, 2.1 months; OS, 12.3 months), compared with those with a low CMI (PFS, 5.8 months; OS, 21.7 months), Dr. Visvanathan and her associates reported (J Clin Oncol. 2016 Nov. 21 doi: 10.1200/JCO.2015.66.2080).
Upon multivariable analysis, a high CMI versus low CMI at 4 weeks was independently associated with poorer PFS (hazard ratio, 1.79; 95% CI, 1.23-2.60; P = .002) and OS (hazard ratio, 1.75; 95% CI, 1.21-2.54; P = .003). Rising CMI levels from baseline to week 4 also were associated with worse PFS (P less than .001) as well as a 4.6-fold increase in the risk of progressive disease at first restaging (OR, 4.58; 95% CI, 1.82-11.60; P = .001) when compared with either a drop in CMI or no change at all.
The CMI level at week 4 was observed to be a strong predictor of PFS and improved the prediction of the base model, even in the presence of circulating tumor cells (P = .004).
Dr. Visvanathan and her colleagues concluded that these findings “must be validated to determine the clinical usefulness of the cMethDNA assay for specific treatments and tumor phenotypes in patients with metastatic disease and early-stage breast cancer.”
The study was supported by the Avon Foundation for Women, the Breast Cancer Research Foundation, Janssen Diagnostics, the Rubenstein Family Fund, the Susan G. Komen Foundation, and National Institutes of Health Grants. Dr. Visvanathan has declared holding patents, royalties, and other intellectual property, and several coauthors also have declared relationships with industry.
A novel panel of DNA methylation markers may aid in predicting survival outcomes in metastatic breast cancer (MBC), according to new findings.
Using a new quantitative assay known as cMethDNA, researchers found that a high cumulative methylation index (CMI) level, as measured with a six-gene panel after beginning a new treatment, was consistently associated with both progression-free survival and overall survival, as well as progressive disease.
Even though these results are encouraging, however, its clinical application is still unknown, they noted.
“Although monitoring for the change in the CMI for treatment response has prognostic usefulness, its clinical usefulness in influencing changes in therapy must now be evaluated formally in randomized clinical trials,” wrote Kala Visvanathan, MD, of Johns Hopkins University, Baltimore, and her colleagues. “Furthermore, whether there is meaningful risk reclassification of patients with the addition of the CMI is worth careful assessment in future larger validation studies.”
Epigenetic alterations are common in human malignancies, and DNA methylation is a form of epigenetic alteration that is heritable during DNA replication. The authors developed a highly sensitive, high-throughput quantitative multiplex methylation-specific polymerase chain reaction assay known as cMethDNA that was able to detect circulating cell-free methylated DNA.
In this study, 10 genes were tested from 141 women with MBC who were beginning a new systemic therapy and who received treatment at participating academic medical centers.
Among patients with a high CMI, the median progression-free survival and overall survival were significantly shorter (PFS, 2.1 months; OS, 12.3 months), compared with those with a low CMI (PFS, 5.8 months; OS, 21.7 months), Dr. Visvanathan and her associates reported (J Clin Oncol. 2016 Nov. 21 doi: 10.1200/JCO.2015.66.2080).
Upon multivariable analysis, a high CMI versus low CMI at 4 weeks was independently associated with poorer PFS (hazard ratio, 1.79; 95% CI, 1.23-2.60; P = .002) and OS (hazard ratio, 1.75; 95% CI, 1.21-2.54; P = .003). Rising CMI levels from baseline to week 4 also were associated with worse PFS (P less than .001) as well as a 4.6-fold increase in the risk of progressive disease at first restaging (OR, 4.58; 95% CI, 1.82-11.60; P = .001) when compared with either a drop in CMI or no change at all.
The CMI level at week 4 was observed to be a strong predictor of PFS and improved the prediction of the base model, even in the presence of circulating tumor cells (P = .004).
Dr. Visvanathan and her colleagues concluded that these findings “must be validated to determine the clinical usefulness of the cMethDNA assay for specific treatments and tumor phenotypes in patients with metastatic disease and early-stage breast cancer.”
The study was supported by the Avon Foundation for Women, the Breast Cancer Research Foundation, Janssen Diagnostics, the Rubenstein Family Fund, the Susan G. Komen Foundation, and National Institutes of Health Grants. Dr. Visvanathan has declared holding patents, royalties, and other intellectual property, and several coauthors also have declared relationships with industry.
FROM THE JOURNAL OF CLINICAL ONCOLOGY
Key clinical point:
Major finding: Among patients with a high cumulative methylation index, the median PFS and OS were significantly shorter (PFS, 2.1 months; OS, 12.3 months), compared with those with a low CMI (PFS, 5.8 months; OS, 21.7 months).
Data source: An experimental study that evaluated 10 genes from samples obtained from 141 metastatic breast cancer patients at varying intervals.
Disclosures: The study was supported by the Avon Foundation for Women, the Breast Cancer Research Foundation, Janssen Diagnostics, the Rubenstein Family Fund, the Susan G. Komen Foundation, and National Institutes of Health Grants. Dr. Visvanathan has declared holding patents, royalties, and other intellectual property, and several coauthors also have declared relationships with industry.