User login
VIDEO: Dr. Roy Herbst talks combinations, evolutions, and innovations in lung cancer
CHICAGO – One of the main take-home messages in lung cancer this year is the efficacy and tolerability demonstrated in combining checkpoint inhibitors for small-cell lung cancer, Dr. Roy Herbst said in an interview at the annual meeting of the American Society of Clinical Oncology. Combining ipilimumab and nivolumab doubled the response rate in patients with progressive small-cell lung cancer, compared with those receiving only nivolumab.
“The evolution of immunotherapy studies continues to show that immunotherapies are here to stay,” Dr. Herbst said. The next step is to expand on how many respond to them. One possibility is to combine an antiangiogenesis agent with a checkpoint inhibitor to drive T cells into the tumor. Dr. Herbst discusses results he presented at the meeting showing safety and tolerability were achieved when combining pembrolizumab and ramucirumab in patients with non–small-cell lung cancer, and the response rate was close to 30%.
In the video interview, Dr. Herbst, professor and chief of medical oncology, Yale Comprehensive Cancer Center, New Haven, Conn., also discusses the evolution of EGFR and ALK inhibitors, a new antibody drug conjugate showing early promise in small cell lung cancer, and the future of liquid biopsies.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
lnikolaides@frontlinemedcom.com
On Twitter @NikolaidesLaura
CHICAGO – One of the main take-home messages in lung cancer this year is the efficacy and tolerability demonstrated in combining checkpoint inhibitors for small-cell lung cancer, Dr. Roy Herbst said in an interview at the annual meeting of the American Society of Clinical Oncology. Combining ipilimumab and nivolumab doubled the response rate in patients with progressive small-cell lung cancer, compared with those receiving only nivolumab.
“The evolution of immunotherapy studies continues to show that immunotherapies are here to stay,” Dr. Herbst said. The next step is to expand on how many respond to them. One possibility is to combine an antiangiogenesis agent with a checkpoint inhibitor to drive T cells into the tumor. Dr. Herbst discusses results he presented at the meeting showing safety and tolerability were achieved when combining pembrolizumab and ramucirumab in patients with non–small-cell lung cancer, and the response rate was close to 30%.
In the video interview, Dr. Herbst, professor and chief of medical oncology, Yale Comprehensive Cancer Center, New Haven, Conn., also discusses the evolution of EGFR and ALK inhibitors, a new antibody drug conjugate showing early promise in small cell lung cancer, and the future of liquid biopsies.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
lnikolaides@frontlinemedcom.com
On Twitter @NikolaidesLaura
CHICAGO – One of the main take-home messages in lung cancer this year is the efficacy and tolerability demonstrated in combining checkpoint inhibitors for small-cell lung cancer, Dr. Roy Herbst said in an interview at the annual meeting of the American Society of Clinical Oncology. Combining ipilimumab and nivolumab doubled the response rate in patients with progressive small-cell lung cancer, compared with those receiving only nivolumab.
“The evolution of immunotherapy studies continues to show that immunotherapies are here to stay,” Dr. Herbst said. The next step is to expand on how many respond to them. One possibility is to combine an antiangiogenesis agent with a checkpoint inhibitor to drive T cells into the tumor. Dr. Herbst discusses results he presented at the meeting showing safety and tolerability were achieved when combining pembrolizumab and ramucirumab in patients with non–small-cell lung cancer, and the response rate was close to 30%.
In the video interview, Dr. Herbst, professor and chief of medical oncology, Yale Comprehensive Cancer Center, New Haven, Conn., also discusses the evolution of EGFR and ALK inhibitors, a new antibody drug conjugate showing early promise in small cell lung cancer, and the future of liquid biopsies.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
lnikolaides@frontlinemedcom.com
On Twitter @NikolaidesLaura
EXPERT ANALYSIS FROM THE 2016 ASCO ANNUAL MEETING
Investigational CDK4/6 inhibitor shows activity, less toxicity
Abemaciclib, a CDK4/6 inhibitor, showed durable clinical activity when given as continuous single-agent therapy to patients with advanced cancer, including breast cancer and non–small-cell lung cancer, according to investigators.
Neutropenia was rarely observed in patients treated with abemaciclib, the toxicity observed in some patients who receive the only Food and Drug Administration–approved CDK4/6 inhibitor, palbociclib.
“Abemaciclib is a small-molecule inhibitor of CDK4 and CDK6 that is structurally distinct from other dual inhibitors (such as palbociclib and ribociclib) and notably exhibits greater selectivity for CDK4 compared with CDK6,” wrote Dr. Amita Patnaik of South Texas Accelerated Research Therapeutics and her associates (Cancer Discov. 2016 May 23. doi: 10.1158/2159-8290.CD-16-0095).
Furthermore, preclinical models indicate that the drug can cross the blood-brain barrier, suggesting potential efficacy against primary and metastatic tumors involving the central nervous system, they said.
A total of 225 patients with various types of advanced cancers were enrolled in this multicohort phase I study (dose escalation, n = 33; single-agent abemaciclib therapy for breast cancer, n = 47; non–small-cell lung cancer, n = 68; glioblastoma, n = 17; melanoma, n = 26; colorectal cancer, n = 15; abemaciclib plus fulvestrant combination therapy for hormone receptor–positive breast cancer, n = 19). Abemaciclib was given orally to all patients.
Neither dose-limiting toxicity nor maximum tolerated dose was reached in patients treated at levels of 50 mg, 100 mg, 150 mg, or 225 mg once daily. The maximum tolerated dose was 200 mg for patients treated with abemaciclib twice daily.
In the single-agent breast cancer cohort, the disease control rate was 81% for hormone receptor–positive (HR-positive) tumors, 33% for HR-negative tumors, 100% for HR-positive HER2-positive tumors, 72% for HR-positive HER2-negative tumors, and 70% overall. The response rate was 31% for HR-positive tumors, 0% for HR-negative tumors, 36% for HR-positive HER2-positive tumors, 28% for HR-positive HER2-negative tumors, and 23% overall.
The overall response rate was 21% for breast cancer patients receiving abemaciclib plus fulvestrant.
Among the 68 patients with non–small-cell lung cancer, 2 had a partial response and 31 had stable disease. Of the 26 patients with melanoma, 1 had a partial response and 6 had stable disease. Of the 17 patients with glioblastoma, 3 had stable disease.
Overall, there were no study-related deaths. Diarrhea, nausea, and fatigue were the most common adverse events; all were reversible.
Neutropenia was observed in 39 patients (23% of 173 patients in the single-agent tumor-specific cohort) – 2 were grade 4 events. Grade 3 neutropenia occurred in 6 patients (32% of 19 patients with HR-positive breast cancer receiving combination therapy with abemaciclib plus fulvestrant).
“Previous reports have identified neutropenia as an adverse event associated with dual inhibition of CDK4 and CDK6. However, abemaciclib given as a single agent on a continuous schedule in the tumor-specific cohorts was associated with an acceptable incidence of investigator-reported grade 3 (9%, 16 of 173 patients) or grade 4 (1%, 2 of 173 patients) neutropenia,” wrote the investigators.
“In summary, the results of this clinical trial demonstrate the safety and antitumor activity of abemaciclib as a single agent and support its further development both as monotherapy and in rational combinations. Furthermore, these findings validate CDK4 and CDK6 as anticancer drug targets and translate preclinical predictions regarding therapeutic targeting of cell-cycle derangements in cancer into clinical efficacy,”they wrote.
Eli Lilly funded the study. Eight investigators reported serving in advisory roles, having ownership or stock interest in, or receiving financial compensation from multiple companies including Eli Lilly.
On Twitter @JessCraig_OP
Abemaciclib, a CDK4/6 inhibitor, showed durable clinical activity when given as continuous single-agent therapy to patients with advanced cancer, including breast cancer and non–small-cell lung cancer, according to investigators.
Neutropenia was rarely observed in patients treated with abemaciclib, the toxicity observed in some patients who receive the only Food and Drug Administration–approved CDK4/6 inhibitor, palbociclib.
“Abemaciclib is a small-molecule inhibitor of CDK4 and CDK6 that is structurally distinct from other dual inhibitors (such as palbociclib and ribociclib) and notably exhibits greater selectivity for CDK4 compared with CDK6,” wrote Dr. Amita Patnaik of South Texas Accelerated Research Therapeutics and her associates (Cancer Discov. 2016 May 23. doi: 10.1158/2159-8290.CD-16-0095).
Furthermore, preclinical models indicate that the drug can cross the blood-brain barrier, suggesting potential efficacy against primary and metastatic tumors involving the central nervous system, they said.
A total of 225 patients with various types of advanced cancers were enrolled in this multicohort phase I study (dose escalation, n = 33; single-agent abemaciclib therapy for breast cancer, n = 47; non–small-cell lung cancer, n = 68; glioblastoma, n = 17; melanoma, n = 26; colorectal cancer, n = 15; abemaciclib plus fulvestrant combination therapy for hormone receptor–positive breast cancer, n = 19). Abemaciclib was given orally to all patients.
Neither dose-limiting toxicity nor maximum tolerated dose was reached in patients treated at levels of 50 mg, 100 mg, 150 mg, or 225 mg once daily. The maximum tolerated dose was 200 mg for patients treated with abemaciclib twice daily.
In the single-agent breast cancer cohort, the disease control rate was 81% for hormone receptor–positive (HR-positive) tumors, 33% for HR-negative tumors, 100% for HR-positive HER2-positive tumors, 72% for HR-positive HER2-negative tumors, and 70% overall. The response rate was 31% for HR-positive tumors, 0% for HR-negative tumors, 36% for HR-positive HER2-positive tumors, 28% for HR-positive HER2-negative tumors, and 23% overall.
The overall response rate was 21% for breast cancer patients receiving abemaciclib plus fulvestrant.
Among the 68 patients with non–small-cell lung cancer, 2 had a partial response and 31 had stable disease. Of the 26 patients with melanoma, 1 had a partial response and 6 had stable disease. Of the 17 patients with glioblastoma, 3 had stable disease.
Overall, there were no study-related deaths. Diarrhea, nausea, and fatigue were the most common adverse events; all were reversible.
Neutropenia was observed in 39 patients (23% of 173 patients in the single-agent tumor-specific cohort) – 2 were grade 4 events. Grade 3 neutropenia occurred in 6 patients (32% of 19 patients with HR-positive breast cancer receiving combination therapy with abemaciclib plus fulvestrant).
“Previous reports have identified neutropenia as an adverse event associated with dual inhibition of CDK4 and CDK6. However, abemaciclib given as a single agent on a continuous schedule in the tumor-specific cohorts was associated with an acceptable incidence of investigator-reported grade 3 (9%, 16 of 173 patients) or grade 4 (1%, 2 of 173 patients) neutropenia,” wrote the investigators.
“In summary, the results of this clinical trial demonstrate the safety and antitumor activity of abemaciclib as a single agent and support its further development both as monotherapy and in rational combinations. Furthermore, these findings validate CDK4 and CDK6 as anticancer drug targets and translate preclinical predictions regarding therapeutic targeting of cell-cycle derangements in cancer into clinical efficacy,”they wrote.
Eli Lilly funded the study. Eight investigators reported serving in advisory roles, having ownership or stock interest in, or receiving financial compensation from multiple companies including Eli Lilly.
On Twitter @JessCraig_OP
Abemaciclib, a CDK4/6 inhibitor, showed durable clinical activity when given as continuous single-agent therapy to patients with advanced cancer, including breast cancer and non–small-cell lung cancer, according to investigators.
Neutropenia was rarely observed in patients treated with abemaciclib, the toxicity observed in some patients who receive the only Food and Drug Administration–approved CDK4/6 inhibitor, palbociclib.
“Abemaciclib is a small-molecule inhibitor of CDK4 and CDK6 that is structurally distinct from other dual inhibitors (such as palbociclib and ribociclib) and notably exhibits greater selectivity for CDK4 compared with CDK6,” wrote Dr. Amita Patnaik of South Texas Accelerated Research Therapeutics and her associates (Cancer Discov. 2016 May 23. doi: 10.1158/2159-8290.CD-16-0095).
Furthermore, preclinical models indicate that the drug can cross the blood-brain barrier, suggesting potential efficacy against primary and metastatic tumors involving the central nervous system, they said.
A total of 225 patients with various types of advanced cancers were enrolled in this multicohort phase I study (dose escalation, n = 33; single-agent abemaciclib therapy for breast cancer, n = 47; non–small-cell lung cancer, n = 68; glioblastoma, n = 17; melanoma, n = 26; colorectal cancer, n = 15; abemaciclib plus fulvestrant combination therapy for hormone receptor–positive breast cancer, n = 19). Abemaciclib was given orally to all patients.
Neither dose-limiting toxicity nor maximum tolerated dose was reached in patients treated at levels of 50 mg, 100 mg, 150 mg, or 225 mg once daily. The maximum tolerated dose was 200 mg for patients treated with abemaciclib twice daily.
In the single-agent breast cancer cohort, the disease control rate was 81% for hormone receptor–positive (HR-positive) tumors, 33% for HR-negative tumors, 100% for HR-positive HER2-positive tumors, 72% for HR-positive HER2-negative tumors, and 70% overall. The response rate was 31% for HR-positive tumors, 0% for HR-negative tumors, 36% for HR-positive HER2-positive tumors, 28% for HR-positive HER2-negative tumors, and 23% overall.
The overall response rate was 21% for breast cancer patients receiving abemaciclib plus fulvestrant.
Among the 68 patients with non–small-cell lung cancer, 2 had a partial response and 31 had stable disease. Of the 26 patients with melanoma, 1 had a partial response and 6 had stable disease. Of the 17 patients with glioblastoma, 3 had stable disease.
Overall, there were no study-related deaths. Diarrhea, nausea, and fatigue were the most common adverse events; all were reversible.
Neutropenia was observed in 39 patients (23% of 173 patients in the single-agent tumor-specific cohort) – 2 were grade 4 events. Grade 3 neutropenia occurred in 6 patients (32% of 19 patients with HR-positive breast cancer receiving combination therapy with abemaciclib plus fulvestrant).
“Previous reports have identified neutropenia as an adverse event associated with dual inhibition of CDK4 and CDK6. However, abemaciclib given as a single agent on a continuous schedule in the tumor-specific cohorts was associated with an acceptable incidence of investigator-reported grade 3 (9%, 16 of 173 patients) or grade 4 (1%, 2 of 173 patients) neutropenia,” wrote the investigators.
“In summary, the results of this clinical trial demonstrate the safety and antitumor activity of abemaciclib as a single agent and support its further development both as monotherapy and in rational combinations. Furthermore, these findings validate CDK4 and CDK6 as anticancer drug targets and translate preclinical predictions regarding therapeutic targeting of cell-cycle derangements in cancer into clinical efficacy,”they wrote.
Eli Lilly funded the study. Eight investigators reported serving in advisory roles, having ownership or stock interest in, or receiving financial compensation from multiple companies including Eli Lilly.
On Twitter @JessCraig_OP
FROM CANCER DISCOVERY
Key clinical point: A phase I trial indicates that abemaciclib is safe and shows activity in treating patients with advanced breast and other cancers.
Major finding: In the single-agent breast cancer cohort, the overall disease control rate was 70%. Incidence of neutropenia was 9% for grade 3 and 1% for grade 4.
Data source: A multicenter phase I dose-escalation and tumor-specific cohort study of 225 patients with advanced cancers.
Disclosures: Eli Lilly funded the study. Eight investigators reported serving in advisory roles, having ownership or stock interest in, or receiving financial compensation from multiple companies including Eli Lilly.
Precision targeting yields better phase I efficacy outcomes
Patients had significantly better outcomes when treatments were selected based on biomarker analysis of their tumors, in a meta-analysis of 346 phase I trials involving 351 study arms and 13,203 cancer patients.
Up to now, the purpose of phase I trials has been to determine safety based on adverse effects and tolerability. “Our analysis really shows that these days it is completely outdated and that with biomarker selection and especially genomic biomarkers, we can reach high response rates even in phase I trials,” Maria Schwaederle, Pharm.D., of the center for personalized cancer therapy at the University of California, San Diego, said in a presscast leading up to the annual meeting of the American Society of Clinical Oncology.
For the meta-analysis, Dr. Schwaederle and her colleagues performed a PubMed search on phase I clinical trials in cancer published in the years 2011-2013. They included studies using a single agent that reported adequate efficacy endpoints for response rate and progression-free survival (PSF). Overall survival was rarely reported and so was excluded.
Personalized therapy was defined as molecular biomarker–based selection of a treatment or at least half the patients in a trial having a specific tumor known to harbor the biomarker. The study compared outcomes based on a personalized strategy with those that were not.
“We think that the results were striking here,” Dr. Schwaederle said. “We can see that personalized therapy that is [based on] biomarker-selected treatments was really associated with significantly higher response rates and progression-free survival.” Multivariable analysis showed that response rates were sixfold higher with the personalized approach (30.6%, n = 58 in the personalized trials, vs. 4.9%, n = 293 in those not personalized; P less than .0001), and median PFS practically doubled (5.7 months, n = 7 in personalized trials vs. 2.95 months, n = 38 in those not personalized; P = .0002).
Most (98.3%) of the personalized arms used targeted agents. However, 76% of the arms using targeted agents did not select patients using a related biomarker and were thus nonpersonalized in their approach to treatment. A subanalysis showed that targeted drugs that were applied in a nontargeted manner had much worse response rates than targeted therapies applied using biomarker-based selection and were equivalent to cytotoxic therapies (personalized approach, 31.1%; nontargeted approach, 5.1%; P less than 0.0001; cytotoxic drugs, 4.7%, P = .63 vs. nonpersonalized approach). The median PFS was also similar for the nonpersonalized and cytotoxic strategies (3.3 vs. 2.5 months, P = .22).
Better targeting with genomic biomarkers
The investigators found that patients selected based on genomic (DNA) biomarkers had almost double the response rates (42%) of patients selected based on protein biomarkers (22.4%, P = .001). The reasons for this difference were not clear. “We might think that the target in the end is a protein, so we would expect maybe to see better results with protein expression,” Dr. Schwaederle said. But she pointed to the example of patients with lung cancer and epidermal growth factor receptor genetic alterations. If looking only for epidermal growth factor receptor protein overexpression, she said, “we wouldn’t see any response, but only the patients that have specific alterations in this gene respond very well.” So in that case, genetic detection appears to be more sensitive than a protein biomarker–based test.
Dr. Schwaederle addressed the question that has often arisen of whether targeted agents are just better therapies. “Our analysis showed that it is not just that the therapies are better but that targeted therapies must be given to the right patients,” she said. “Indeed, when targeted therapies were given to patients without a biomarker selection, the response rates were only about 5%.”
The response rate in the range of 40% using genetic biomarkers in these phase I trials suggests that incorporating such targeted approaches even at this early stage of drug testing may potentially yield useful information on efficacy.
Dr. Don Dizon, presscast moderator and chair of ASCO’s Cancer Communications Committee, said that precision medicine “is here and that we can use patient selection using either changes in a tumor’s DNA, called genomic changes, or even protein biomarkers and do much better than we have done in the past.”
Dr. Schwaederle noted that one limitation of her study is that more recent trials, unpublished at the time of her analysis, could influence the results today.
The study received funding from the Joan and Irwin Jacobs Philanthropic Fund. Two coauthors reported extensive financial relationships with pharmaceutical or other commercial and noncommercial entities.
Patients had significantly better outcomes when treatments were selected based on biomarker analysis of their tumors, in a meta-analysis of 346 phase I trials involving 351 study arms and 13,203 cancer patients.
Up to now, the purpose of phase I trials has been to determine safety based on adverse effects and tolerability. “Our analysis really shows that these days it is completely outdated and that with biomarker selection and especially genomic biomarkers, we can reach high response rates even in phase I trials,” Maria Schwaederle, Pharm.D., of the center for personalized cancer therapy at the University of California, San Diego, said in a presscast leading up to the annual meeting of the American Society of Clinical Oncology.
For the meta-analysis, Dr. Schwaederle and her colleagues performed a PubMed search on phase I clinical trials in cancer published in the years 2011-2013. They included studies using a single agent that reported adequate efficacy endpoints for response rate and progression-free survival (PSF). Overall survival was rarely reported and so was excluded.
Personalized therapy was defined as molecular biomarker–based selection of a treatment or at least half the patients in a trial having a specific tumor known to harbor the biomarker. The study compared outcomes based on a personalized strategy with those that were not.
“We think that the results were striking here,” Dr. Schwaederle said. “We can see that personalized therapy that is [based on] biomarker-selected treatments was really associated with significantly higher response rates and progression-free survival.” Multivariable analysis showed that response rates were sixfold higher with the personalized approach (30.6%, n = 58 in the personalized trials, vs. 4.9%, n = 293 in those not personalized; P less than .0001), and median PFS practically doubled (5.7 months, n = 7 in personalized trials vs. 2.95 months, n = 38 in those not personalized; P = .0002).
Most (98.3%) of the personalized arms used targeted agents. However, 76% of the arms using targeted agents did not select patients using a related biomarker and were thus nonpersonalized in their approach to treatment. A subanalysis showed that targeted drugs that were applied in a nontargeted manner had much worse response rates than targeted therapies applied using biomarker-based selection and were equivalent to cytotoxic therapies (personalized approach, 31.1%; nontargeted approach, 5.1%; P less than 0.0001; cytotoxic drugs, 4.7%, P = .63 vs. nonpersonalized approach). The median PFS was also similar for the nonpersonalized and cytotoxic strategies (3.3 vs. 2.5 months, P = .22).
Better targeting with genomic biomarkers
The investigators found that patients selected based on genomic (DNA) biomarkers had almost double the response rates (42%) of patients selected based on protein biomarkers (22.4%, P = .001). The reasons for this difference were not clear. “We might think that the target in the end is a protein, so we would expect maybe to see better results with protein expression,” Dr. Schwaederle said. But she pointed to the example of patients with lung cancer and epidermal growth factor receptor genetic alterations. If looking only for epidermal growth factor receptor protein overexpression, she said, “we wouldn’t see any response, but only the patients that have specific alterations in this gene respond very well.” So in that case, genetic detection appears to be more sensitive than a protein biomarker–based test.
Dr. Schwaederle addressed the question that has often arisen of whether targeted agents are just better therapies. “Our analysis showed that it is not just that the therapies are better but that targeted therapies must be given to the right patients,” she said. “Indeed, when targeted therapies were given to patients without a biomarker selection, the response rates were only about 5%.”
The response rate in the range of 40% using genetic biomarkers in these phase I trials suggests that incorporating such targeted approaches even at this early stage of drug testing may potentially yield useful information on efficacy.
Dr. Don Dizon, presscast moderator and chair of ASCO’s Cancer Communications Committee, said that precision medicine “is here and that we can use patient selection using either changes in a tumor’s DNA, called genomic changes, or even protein biomarkers and do much better than we have done in the past.”
Dr. Schwaederle noted that one limitation of her study is that more recent trials, unpublished at the time of her analysis, could influence the results today.
The study received funding from the Joan and Irwin Jacobs Philanthropic Fund. Two coauthors reported extensive financial relationships with pharmaceutical or other commercial and noncommercial entities.
Patients had significantly better outcomes when treatments were selected based on biomarker analysis of their tumors, in a meta-analysis of 346 phase I trials involving 351 study arms and 13,203 cancer patients.
Up to now, the purpose of phase I trials has been to determine safety based on adverse effects and tolerability. “Our analysis really shows that these days it is completely outdated and that with biomarker selection and especially genomic biomarkers, we can reach high response rates even in phase I trials,” Maria Schwaederle, Pharm.D., of the center for personalized cancer therapy at the University of California, San Diego, said in a presscast leading up to the annual meeting of the American Society of Clinical Oncology.
For the meta-analysis, Dr. Schwaederle and her colleagues performed a PubMed search on phase I clinical trials in cancer published in the years 2011-2013. They included studies using a single agent that reported adequate efficacy endpoints for response rate and progression-free survival (PSF). Overall survival was rarely reported and so was excluded.
Personalized therapy was defined as molecular biomarker–based selection of a treatment or at least half the patients in a trial having a specific tumor known to harbor the biomarker. The study compared outcomes based on a personalized strategy with those that were not.
“We think that the results were striking here,” Dr. Schwaederle said. “We can see that personalized therapy that is [based on] biomarker-selected treatments was really associated with significantly higher response rates and progression-free survival.” Multivariable analysis showed that response rates were sixfold higher with the personalized approach (30.6%, n = 58 in the personalized trials, vs. 4.9%, n = 293 in those not personalized; P less than .0001), and median PFS practically doubled (5.7 months, n = 7 in personalized trials vs. 2.95 months, n = 38 in those not personalized; P = .0002).
Most (98.3%) of the personalized arms used targeted agents. However, 76% of the arms using targeted agents did not select patients using a related biomarker and were thus nonpersonalized in their approach to treatment. A subanalysis showed that targeted drugs that were applied in a nontargeted manner had much worse response rates than targeted therapies applied using biomarker-based selection and were equivalent to cytotoxic therapies (personalized approach, 31.1%; nontargeted approach, 5.1%; P less than 0.0001; cytotoxic drugs, 4.7%, P = .63 vs. nonpersonalized approach). The median PFS was also similar for the nonpersonalized and cytotoxic strategies (3.3 vs. 2.5 months, P = .22).
Better targeting with genomic biomarkers
The investigators found that patients selected based on genomic (DNA) biomarkers had almost double the response rates (42%) of patients selected based on protein biomarkers (22.4%, P = .001). The reasons for this difference were not clear. “We might think that the target in the end is a protein, so we would expect maybe to see better results with protein expression,” Dr. Schwaederle said. But she pointed to the example of patients with lung cancer and epidermal growth factor receptor genetic alterations. If looking only for epidermal growth factor receptor protein overexpression, she said, “we wouldn’t see any response, but only the patients that have specific alterations in this gene respond very well.” So in that case, genetic detection appears to be more sensitive than a protein biomarker–based test.
Dr. Schwaederle addressed the question that has often arisen of whether targeted agents are just better therapies. “Our analysis showed that it is not just that the therapies are better but that targeted therapies must be given to the right patients,” she said. “Indeed, when targeted therapies were given to patients without a biomarker selection, the response rates were only about 5%.”
The response rate in the range of 40% using genetic biomarkers in these phase I trials suggests that incorporating such targeted approaches even at this early stage of drug testing may potentially yield useful information on efficacy.
Dr. Don Dizon, presscast moderator and chair of ASCO’s Cancer Communications Committee, said that precision medicine “is here and that we can use patient selection using either changes in a tumor’s DNA, called genomic changes, or even protein biomarkers and do much better than we have done in the past.”
Dr. Schwaederle noted that one limitation of her study is that more recent trials, unpublished at the time of her analysis, could influence the results today.
The study received funding from the Joan and Irwin Jacobs Philanthropic Fund. Two coauthors reported extensive financial relationships with pharmaceutical or other commercial and noncommercial entities.
FROM THE 2016 ASCO ANNUAL MEETING
Key clinical point: Precision drug targeting in phase I trials may yield efficacy information.
Major finding: Genomic targeting yielded a 42% response rate vs. 5% without targeting.
Data source: A meta-analysis of 346 phase I trials with 351 treatment arms and 13,203 cancer patients.
Disclosures: The study received funding from the Joan and Irwin Jacobs Philanthropic Fund. Two coauthors reported extensive financial relationships with pharmaceutical or other commercial and noncommercial entities.
Smoking linked to longer hospital stay
Current and former cigarette smoking were associated with a longer hospital stay for patients with lung cancer, chronic obstructive pulmonary disease, or ischemic heart disease, according to a report published in Hospital Practice.
Researchers assessed factors influencing hospital length of stay (LOS) as a first step toward shortening that measure, improving inpatient services, and cutting hospital costs in Iran, where this issue has not been studied previously. The overall prevalence of smoking in Iran is 12.5%, and it is much more common among men than women, said Satar Rezaei, Ph.D., of the Research Centre for Environmental Determinants of Health, Kermanshah (Iran) University of Medical Sciences, and his associates.
They assessed LOS in 1,271 patients aged 35-93 years (mean age, 63 years) who were hospitalized at 11 Tehran medical centers during a 1-year period. Slightly more than half (54%) of these patients had never smoked, while 34% were current smokers and 12% were former smokers. A total of 415 (32%) had lung cancer, 427 (34%) had COPD, and 429 (34%) had ischemic heart disease. The overall mean LOS was 7.3 days, with a range from 1 to 45 days.
The mean LOS was 48% longer among current smokers and 15% longer among former smokers than among nonsmokers. It was 9.4 days for current smokers and 7.3 days for former smokers, compared with 6.0 days for nonsmokers, Dr. Rezaei and his associates said (Hosp. Pract. 2016. doi: 10.1080/21548331.2016.1178579).
Patient cigarette smoking imposes a heavy financial burden on these hospitals and contributes to their shortage of resources. Hospital-based smoking cessation programs would be useful in shortening LOS and sparing hospital resources, the researchers added.
This study was supported by Tehran University of Medical Sciences. Dr. Rezaei and his associates reported having no relevant financial disclosures.
Current and former cigarette smoking were associated with a longer hospital stay for patients with lung cancer, chronic obstructive pulmonary disease, or ischemic heart disease, according to a report published in Hospital Practice.
Researchers assessed factors influencing hospital length of stay (LOS) as a first step toward shortening that measure, improving inpatient services, and cutting hospital costs in Iran, where this issue has not been studied previously. The overall prevalence of smoking in Iran is 12.5%, and it is much more common among men than women, said Satar Rezaei, Ph.D., of the Research Centre for Environmental Determinants of Health, Kermanshah (Iran) University of Medical Sciences, and his associates.
They assessed LOS in 1,271 patients aged 35-93 years (mean age, 63 years) who were hospitalized at 11 Tehran medical centers during a 1-year period. Slightly more than half (54%) of these patients had never smoked, while 34% were current smokers and 12% were former smokers. A total of 415 (32%) had lung cancer, 427 (34%) had COPD, and 429 (34%) had ischemic heart disease. The overall mean LOS was 7.3 days, with a range from 1 to 45 days.
The mean LOS was 48% longer among current smokers and 15% longer among former smokers than among nonsmokers. It was 9.4 days for current smokers and 7.3 days for former smokers, compared with 6.0 days for nonsmokers, Dr. Rezaei and his associates said (Hosp. Pract. 2016. doi: 10.1080/21548331.2016.1178579).
Patient cigarette smoking imposes a heavy financial burden on these hospitals and contributes to their shortage of resources. Hospital-based smoking cessation programs would be useful in shortening LOS and sparing hospital resources, the researchers added.
This study was supported by Tehran University of Medical Sciences. Dr. Rezaei and his associates reported having no relevant financial disclosures.
Current and former cigarette smoking were associated with a longer hospital stay for patients with lung cancer, chronic obstructive pulmonary disease, or ischemic heart disease, according to a report published in Hospital Practice.
Researchers assessed factors influencing hospital length of stay (LOS) as a first step toward shortening that measure, improving inpatient services, and cutting hospital costs in Iran, where this issue has not been studied previously. The overall prevalence of smoking in Iran is 12.5%, and it is much more common among men than women, said Satar Rezaei, Ph.D., of the Research Centre for Environmental Determinants of Health, Kermanshah (Iran) University of Medical Sciences, and his associates.
They assessed LOS in 1,271 patients aged 35-93 years (mean age, 63 years) who were hospitalized at 11 Tehran medical centers during a 1-year period. Slightly more than half (54%) of these patients had never smoked, while 34% were current smokers and 12% were former smokers. A total of 415 (32%) had lung cancer, 427 (34%) had COPD, and 429 (34%) had ischemic heart disease. The overall mean LOS was 7.3 days, with a range from 1 to 45 days.
The mean LOS was 48% longer among current smokers and 15% longer among former smokers than among nonsmokers. It was 9.4 days for current smokers and 7.3 days for former smokers, compared with 6.0 days for nonsmokers, Dr. Rezaei and his associates said (Hosp. Pract. 2016. doi: 10.1080/21548331.2016.1178579).
Patient cigarette smoking imposes a heavy financial burden on these hospitals and contributes to their shortage of resources. Hospital-based smoking cessation programs would be useful in shortening LOS and sparing hospital resources, the researchers added.
This study was supported by Tehran University of Medical Sciences. Dr. Rezaei and his associates reported having no relevant financial disclosures.
FROM HOSPITAL PRACTICE
Key clinical point: Current and former cigarette smoking are associated with a longer hospital stay for patients with lung cancer, chronic obstructive pulmonary disease, or ischemic heart disease.
Major finding: The mean LOS was 48% longer among current smokers and 15% longer among former smokers than among nonsmokers.
Data source: A retrospective cohort study involving 1,271 patients hospitalized in Iran during a 1-year period.
Disclosures: This study was supported by Tehran University of Medical Sciences. Dr. Rezaei and his associates reported having no relevant financial disclosures.
VIDEO: Wedge resection beats SBRT for stage I lung cancer treatment
BALTIMORE – Surgical resection of early-stage non–small cell lung cancer afforded a superior survival advantage for patients than stereotactic body radiation therapy (SBRT), according to a study presented at the 2016 annual meeting of the American Association for Thoracic Surgery.
While an increasing number of non–small cell lung cancer patients have been treated with SBRT, it appears that surgery may still be the better option. Analysis of both matched and unmatched patient groups found that SBRT was associated with significantly lower survival than wedge resection.
“Frankly, I was surprised to see such a big difference between SBRT and wedge resection,” said Dr. Walter Weder, professor of surgery at University Hospital Zürich, in an interview at AATS 2016. Dr Weder served as a discussant on the paper, and said the results confirm that surgeons should be involved in discussions with patients when they are considering treatment options. “Surgery can be done safely... and patients should know this information.”
Dr. Weder reported no relevant financial disclosures.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
On Twitter @richpizzi
BALTIMORE – Surgical resection of early-stage non–small cell lung cancer afforded a superior survival advantage for patients than stereotactic body radiation therapy (SBRT), according to a study presented at the 2016 annual meeting of the American Association for Thoracic Surgery.
While an increasing number of non–small cell lung cancer patients have been treated with SBRT, it appears that surgery may still be the better option. Analysis of both matched and unmatched patient groups found that SBRT was associated with significantly lower survival than wedge resection.
“Frankly, I was surprised to see such a big difference between SBRT and wedge resection,” said Dr. Walter Weder, professor of surgery at University Hospital Zürich, in an interview at AATS 2016. Dr Weder served as a discussant on the paper, and said the results confirm that surgeons should be involved in discussions with patients when they are considering treatment options. “Surgery can be done safely... and patients should know this information.”
Dr. Weder reported no relevant financial disclosures.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
On Twitter @richpizzi
BALTIMORE – Surgical resection of early-stage non–small cell lung cancer afforded a superior survival advantage for patients than stereotactic body radiation therapy (SBRT), according to a study presented at the 2016 annual meeting of the American Association for Thoracic Surgery.
While an increasing number of non–small cell lung cancer patients have been treated with SBRT, it appears that surgery may still be the better option. Analysis of both matched and unmatched patient groups found that SBRT was associated with significantly lower survival than wedge resection.
“Frankly, I was surprised to see such a big difference between SBRT and wedge resection,” said Dr. Walter Weder, professor of surgery at University Hospital Zürich, in an interview at AATS 2016. Dr Weder served as a discussant on the paper, and said the results confirm that surgeons should be involved in discussions with patients when they are considering treatment options. “Surgery can be done safely... and patients should know this information.”
Dr. Weder reported no relevant financial disclosures.
The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
On Twitter @richpizzi
AT AATS 2016
Early palliative care for cancer patients benefits caregivers
Introducing palliative care in combination with standard oncology care immediately following a cancer diagnosis results in improved quality of life and lower incidence of depression for caregivers of cancer patients.
“The integration of palliative care can improve patient care but the evidence is lacking about whether or not there are benefits [for] caregivers,” Dr. Areej El-Jawahri of Massachusetts General Hospital, Boston, said in a presscast leading up to the annual meeting of the American Society of Clinical Oncology.
“This study suggests that early palliative care creates a powerful positive feedback loop in families facing cancer. While patients receive a direct benefit from early palliative care, their caregivers experience a positive downstream effect, which may make it easier for them to care for their loved ones,” she said.
Investigators enrolled 275 family caregivers of patients newly diagnosed with incurable lung or gastrointestinal cancers. Patients were randomly assigned to receive early palliative care in addition to standard oncology care or to receive standard oncology care alone.
Palliative care involved a multifaceted team including nurses, social workers, and psychologists. The palliative care intervention was patient focused, and caregivers, who were defined as a relative or friend identified by the patient as the primary caregiver, were not required to attend palliative care appointments. However, about 50% of caregivers did attend, according to Dr. El-Jawahri.
At time of enrollment and then at time points 12 and 14 weeks post enrollment, caregivers completed standard questionnaires, the 36-Item Short Form Health Survey and the Hospital Anxiety and Depression Scale, that assessed quality of life and mood.
Twelve weeks after the cancer diagnosis, caregivers who received early palliative care reported significantly lower depression symptoms while vitality and social functioning improved. For patients who did not receive early palliative care, their caregivers’ vitality and social functioning decreased.
“This is the first study showing a positive impact of a patient-focused palliative care intervention on family caregivers,” said Dr. El-Jawahri.
“This study really points out that we have so many ways to help our patients and their families,” Dr. Julie Vose, president of ASCO, said during the presscast.
On Twitter @JessCraig_OP
Introducing palliative care in combination with standard oncology care immediately following a cancer diagnosis results in improved quality of life and lower incidence of depression for caregivers of cancer patients.
“The integration of palliative care can improve patient care but the evidence is lacking about whether or not there are benefits [for] caregivers,” Dr. Areej El-Jawahri of Massachusetts General Hospital, Boston, said in a presscast leading up to the annual meeting of the American Society of Clinical Oncology.
“This study suggests that early palliative care creates a powerful positive feedback loop in families facing cancer. While patients receive a direct benefit from early palliative care, their caregivers experience a positive downstream effect, which may make it easier for them to care for their loved ones,” she said.
Investigators enrolled 275 family caregivers of patients newly diagnosed with incurable lung or gastrointestinal cancers. Patients were randomly assigned to receive early palliative care in addition to standard oncology care or to receive standard oncology care alone.
Palliative care involved a multifaceted team including nurses, social workers, and psychologists. The palliative care intervention was patient focused, and caregivers, who were defined as a relative or friend identified by the patient as the primary caregiver, were not required to attend palliative care appointments. However, about 50% of caregivers did attend, according to Dr. El-Jawahri.
At time of enrollment and then at time points 12 and 14 weeks post enrollment, caregivers completed standard questionnaires, the 36-Item Short Form Health Survey and the Hospital Anxiety and Depression Scale, that assessed quality of life and mood.
Twelve weeks after the cancer diagnosis, caregivers who received early palliative care reported significantly lower depression symptoms while vitality and social functioning improved. For patients who did not receive early palliative care, their caregivers’ vitality and social functioning decreased.
“This is the first study showing a positive impact of a patient-focused palliative care intervention on family caregivers,” said Dr. El-Jawahri.
“This study really points out that we have so many ways to help our patients and their families,” Dr. Julie Vose, president of ASCO, said during the presscast.
On Twitter @JessCraig_OP
Introducing palliative care in combination with standard oncology care immediately following a cancer diagnosis results in improved quality of life and lower incidence of depression for caregivers of cancer patients.
“The integration of palliative care can improve patient care but the evidence is lacking about whether or not there are benefits [for] caregivers,” Dr. Areej El-Jawahri of Massachusetts General Hospital, Boston, said in a presscast leading up to the annual meeting of the American Society of Clinical Oncology.
“This study suggests that early palliative care creates a powerful positive feedback loop in families facing cancer. While patients receive a direct benefit from early palliative care, their caregivers experience a positive downstream effect, which may make it easier for them to care for their loved ones,” she said.
Investigators enrolled 275 family caregivers of patients newly diagnosed with incurable lung or gastrointestinal cancers. Patients were randomly assigned to receive early palliative care in addition to standard oncology care or to receive standard oncology care alone.
Palliative care involved a multifaceted team including nurses, social workers, and psychologists. The palliative care intervention was patient focused, and caregivers, who were defined as a relative or friend identified by the patient as the primary caregiver, were not required to attend palliative care appointments. However, about 50% of caregivers did attend, according to Dr. El-Jawahri.
At time of enrollment and then at time points 12 and 14 weeks post enrollment, caregivers completed standard questionnaires, the 36-Item Short Form Health Survey and the Hospital Anxiety and Depression Scale, that assessed quality of life and mood.
Twelve weeks after the cancer diagnosis, caregivers who received early palliative care reported significantly lower depression symptoms while vitality and social functioning improved. For patients who did not receive early palliative care, their caregivers’ vitality and social functioning decreased.
“This is the first study showing a positive impact of a patient-focused palliative care intervention on family caregivers,” said Dr. El-Jawahri.
“This study really points out that we have so many ways to help our patients and their families,” Dr. Julie Vose, president of ASCO, said during the presscast.
On Twitter @JessCraig_OP
FROM THE 2016 ASCO ANNUAL MEETING
Key clinical point: Early palliative care for cancer patients improved quality of life and lowered the incidence of depression for caregivers.
Major finding: After 12 weeks, caregivers of patients who received early palliative care reported significantly lower depression symptoms. Vitality and social functioning also improved.
Data source: A randomized clinical trial of 275 family caregivers of patients newly diagnosed with incurable lung or gastrointestinal cancers.
Disclosures: The National Institutes of Health funded the study. Three of the investigators reported serving in advisory roles or receiving financial compensation or honoraria from several companies.
FDG-PET/CT leads pack for small-cell lung cancer staging
For pretreatment staging of small-cell lung cancer (SCLC) the use of positron-emission tomography combined with CT was more sensitive compared with several other tests, according to a new report on a review of studies.
Overall, positron emission tomography using [F]-fluorodeoxyglucose as a radiotracer combined with CT (FDG-PET/CT) had greater sensitivity to detect osseous metastases than did bone scintigraphy or CT alone, according to Dr. Jonathan R. Treadwell, Ph.D., of ECRI Institute–Penn Medicine’s Evidence-based Practice Center in Plymouth Meeting, Pa., and colleagues. In addition, the researchers concluded that adding FDG-PET/CT to the protocol for patients who have undergone standard staging increased the sensitivity for detecting additional metastases. Data on endobronchial ultrasound were insufficient to draw any conclusions.
The findings generally line up with recent guidelines from the American College of Radiology (ACR) and American College of Chest Physicians (ACCP). In 2014, the ACR gave the highest rating of “usually appropriate” (with regard to staging SCLC) to FDG-PET/CT from skull base to mid-thigh, while bone scintigraphy was rated as “may be appropriate” and not necessary if PET/CT had been done, the researchers wrote. The 2013 ACCP guideline “suggested” FDG PET instead of bone scintigraphy for patients with limited disease, they added.
The researchers reviewed data from seven studies to assess the accuracy and effectiveness of several imaging modalities for the pretreatment staging of SCLC. The report was generated for the Agency for Healthcare Research and Quality (AHRQ) as part of its Comparative Effectiveness Review series, and is not an official AHRQ position, the researchers noted.
Combining FDG-PET with CT scanning has demonstrated even greater effectiveness at identifying malignant tumors and metabolically active metastases than has PET alone, because the CT allows for more localized anatomic detail, the researchers explained. “False negative scans usually result from non–metabolically active sites of tumor or from suboptimal quality studies,” they said, while false positives using FDG-PET are usually attributed to inflammation or metabolically active infection.
The meta-analysis included data on endobronchial ultrasound, which involves ultrasound to view structures inside and adjacent to the airway; bone scintigraphy, a less expensive planar molecular imaging technique; and CT alone.
Comparative evidence on pretreatment staging for SCLC is limited, according to the researchers. The data did not allow them to determine how FDG-PET/CT compared to other imaging in terms of specificity, and any type of imaging can yield false positives, they said. However, higher sensitivity alone can benefit patients in terms of improving patient selection for optimal therapy, sparing patients chemotherapy if not needed, and improving the prediction value of ongoing research, they noted.
“Although high-quality evidence may not be voluminous, I think most physicians would agree with the conclusion that a bone scan is not mandatory in the work-up of possible SCLC, if a PET/CT has been done,” Dr. W. Michael Alberts of the Moffitt Cancer Center in Tampa, Fla., said in an interview.
Cost might play a role in why the guidelines are being issued at this time, he noted, because “the initial work-up of the patient with suspected SCLC may prove to be quite expensive, and the elimination of a superfluous test may be a fiscal winner.” However, more research is needed in this area, particularly in the areas of including the order of pretreatment testing and the incorporation of new procedures and imaging modalities, he added. “Perhaps more intellectually challenging, however, might be the question of why SCLC is becoming less common, or why has improvement in treatment been so slow compared to NSCLC,” he added.
The researchers had no financial conflicts to disclose.
For pretreatment staging of small-cell lung cancer (SCLC) the use of positron-emission tomography combined with CT was more sensitive compared with several other tests, according to a new report on a review of studies.
Overall, positron emission tomography using [F]-fluorodeoxyglucose as a radiotracer combined with CT (FDG-PET/CT) had greater sensitivity to detect osseous metastases than did bone scintigraphy or CT alone, according to Dr. Jonathan R. Treadwell, Ph.D., of ECRI Institute–Penn Medicine’s Evidence-based Practice Center in Plymouth Meeting, Pa., and colleagues. In addition, the researchers concluded that adding FDG-PET/CT to the protocol for patients who have undergone standard staging increased the sensitivity for detecting additional metastases. Data on endobronchial ultrasound were insufficient to draw any conclusions.
The findings generally line up with recent guidelines from the American College of Radiology (ACR) and American College of Chest Physicians (ACCP). In 2014, the ACR gave the highest rating of “usually appropriate” (with regard to staging SCLC) to FDG-PET/CT from skull base to mid-thigh, while bone scintigraphy was rated as “may be appropriate” and not necessary if PET/CT had been done, the researchers wrote. The 2013 ACCP guideline “suggested” FDG PET instead of bone scintigraphy for patients with limited disease, they added.
The researchers reviewed data from seven studies to assess the accuracy and effectiveness of several imaging modalities for the pretreatment staging of SCLC. The report was generated for the Agency for Healthcare Research and Quality (AHRQ) as part of its Comparative Effectiveness Review series, and is not an official AHRQ position, the researchers noted.
Combining FDG-PET with CT scanning has demonstrated even greater effectiveness at identifying malignant tumors and metabolically active metastases than has PET alone, because the CT allows for more localized anatomic detail, the researchers explained. “False negative scans usually result from non–metabolically active sites of tumor or from suboptimal quality studies,” they said, while false positives using FDG-PET are usually attributed to inflammation or metabolically active infection.
The meta-analysis included data on endobronchial ultrasound, which involves ultrasound to view structures inside and adjacent to the airway; bone scintigraphy, a less expensive planar molecular imaging technique; and CT alone.
Comparative evidence on pretreatment staging for SCLC is limited, according to the researchers. The data did not allow them to determine how FDG-PET/CT compared to other imaging in terms of specificity, and any type of imaging can yield false positives, they said. However, higher sensitivity alone can benefit patients in terms of improving patient selection for optimal therapy, sparing patients chemotherapy if not needed, and improving the prediction value of ongoing research, they noted.
“Although high-quality evidence may not be voluminous, I think most physicians would agree with the conclusion that a bone scan is not mandatory in the work-up of possible SCLC, if a PET/CT has been done,” Dr. W. Michael Alberts of the Moffitt Cancer Center in Tampa, Fla., said in an interview.
Cost might play a role in why the guidelines are being issued at this time, he noted, because “the initial work-up of the patient with suspected SCLC may prove to be quite expensive, and the elimination of a superfluous test may be a fiscal winner.” However, more research is needed in this area, particularly in the areas of including the order of pretreatment testing and the incorporation of new procedures and imaging modalities, he added. “Perhaps more intellectually challenging, however, might be the question of why SCLC is becoming less common, or why has improvement in treatment been so slow compared to NSCLC,” he added.
The researchers had no financial conflicts to disclose.
For pretreatment staging of small-cell lung cancer (SCLC) the use of positron-emission tomography combined with CT was more sensitive compared with several other tests, according to a new report on a review of studies.
Overall, positron emission tomography using [F]-fluorodeoxyglucose as a radiotracer combined with CT (FDG-PET/CT) had greater sensitivity to detect osseous metastases than did bone scintigraphy or CT alone, according to Dr. Jonathan R. Treadwell, Ph.D., of ECRI Institute–Penn Medicine’s Evidence-based Practice Center in Plymouth Meeting, Pa., and colleagues. In addition, the researchers concluded that adding FDG-PET/CT to the protocol for patients who have undergone standard staging increased the sensitivity for detecting additional metastases. Data on endobronchial ultrasound were insufficient to draw any conclusions.
The findings generally line up with recent guidelines from the American College of Radiology (ACR) and American College of Chest Physicians (ACCP). In 2014, the ACR gave the highest rating of “usually appropriate” (with regard to staging SCLC) to FDG-PET/CT from skull base to mid-thigh, while bone scintigraphy was rated as “may be appropriate” and not necessary if PET/CT had been done, the researchers wrote. The 2013 ACCP guideline “suggested” FDG PET instead of bone scintigraphy for patients with limited disease, they added.
The researchers reviewed data from seven studies to assess the accuracy and effectiveness of several imaging modalities for the pretreatment staging of SCLC. The report was generated for the Agency for Healthcare Research and Quality (AHRQ) as part of its Comparative Effectiveness Review series, and is not an official AHRQ position, the researchers noted.
Combining FDG-PET with CT scanning has demonstrated even greater effectiveness at identifying malignant tumors and metabolically active metastases than has PET alone, because the CT allows for more localized anatomic detail, the researchers explained. “False negative scans usually result from non–metabolically active sites of tumor or from suboptimal quality studies,” they said, while false positives using FDG-PET are usually attributed to inflammation or metabolically active infection.
The meta-analysis included data on endobronchial ultrasound, which involves ultrasound to view structures inside and adjacent to the airway; bone scintigraphy, a less expensive planar molecular imaging technique; and CT alone.
Comparative evidence on pretreatment staging for SCLC is limited, according to the researchers. The data did not allow them to determine how FDG-PET/CT compared to other imaging in terms of specificity, and any type of imaging can yield false positives, they said. However, higher sensitivity alone can benefit patients in terms of improving patient selection for optimal therapy, sparing patients chemotherapy if not needed, and improving the prediction value of ongoing research, they noted.
“Although high-quality evidence may not be voluminous, I think most physicians would agree with the conclusion that a bone scan is not mandatory in the work-up of possible SCLC, if a PET/CT has been done,” Dr. W. Michael Alberts of the Moffitt Cancer Center in Tampa, Fla., said in an interview.
Cost might play a role in why the guidelines are being issued at this time, he noted, because “the initial work-up of the patient with suspected SCLC may prove to be quite expensive, and the elimination of a superfluous test may be a fiscal winner.” However, more research is needed in this area, particularly in the areas of including the order of pretreatment testing and the incorporation of new procedures and imaging modalities, he added. “Perhaps more intellectually challenging, however, might be the question of why SCLC is becoming less common, or why has improvement in treatment been so slow compared to NSCLC,” he added.
The researchers had no financial conflicts to disclose.
New dose-determining approach applied to pembrolizumab
A 2-mg/kg dosage of pembrolizumab given to adult patients with non–small-cell lung cancer (NSCLC) every 3 weeks is optimal to both reduce tumor size and avoid treatment-related adverse events, according to a new method of analysis.
Current methods for determining the maximum tolerated dose (MTD) based on dose-limiting toxicities may be outmoded for targeted therapies and immunotherapies, where the biologically effective dose (BED) may be much lower than the MTD.
“Using the MTD rather than the BED could expose patients to a higher dose than that necessary to achieve clinical effect and may increase toxicity, which could lower overall clinical effectiveness. Therefore, dose determination in oncology should use a multifactorial approach that includes not only clinical data from the first treatment cycle but extended clinical data, preclinical models, pharmacokinetics, pharmacodynamics, and integrated modeling and simulation,” said Dr. Manash Chatterjee of Merck, and associates (Ann Oncol. 2016 April 26. doi: 10.1093/annonc/mdw174).
Dr. Chatterjee and associates analyzed data from KEYNOTE-001 using various statistical and mathematical modeling techniques to determine the lowest effective drug dosage of pembrolizumab, a highly selective humanized IgG4 monoclonal antibody that targets the programmed death-1 receptor, for treatment of patients with NSCLC.
Patients enrolled in the KEYNOTE-001 trial received pembrolizumab at doses of 2 mg/kg every 3 weeks, 10 mg/kg every 3 weeks, or 10 mg/kg every 2 weeks until disease progression or intolerable toxicity, or until the patient or investigator decided to stop treatment. Sixty-one patients were treated with pembrolizumab at 2 mg/kg every 3 weeks, 287 patients were treated at 10 mg/kg every 3 weeks, and 202 patients were treated at 10 mg/kg every 2 weeks. PD-1 expression was assessed from biopsy samples using a prototype assay.
Decreases from baseline tumor size were observed for 67% of patients with known PD-L1 expression treated with pembrolizumab at 2 mg/kg every 3 weeks, 66% for patients treated at 10 mg/kg every 3 weeks, and 63% for patients treated at 10 mg/kg every 2 weeks.
Data on occurrence and severity of adverse events were collected throughout the study and up to 60 days after treatment was discontinued. Adverse events were graded according to the National Cancer Institute’s Common Terminology Criteria for Adverse Events (version 4.0). Treatment-related adverse events were reported for 47% of patients treated at 2 mg/kg every 3 weeks. The frequency and severity of adverse events were similar for all dosages, the investigators reported.
Exposure-efficacy was evaluated by way of exploratory regression analyses followed by nonlinear mixed effects modeling. The model found no significant difference in the exposure-efficacy relationship based on dosage. PD-L1 expression and EGFR mutation were the only two factors that explained a significant portion of interindividual variability in tumor size decrease.
Exposure-safety was evaluated via logistic regression modeling where safety was defined as the frequency of adverse events. The model revealed that there was no significant relationship between adverse events and exposure to pembrolizumab or dosage. Overall duration of treatment was the only factor that was significantly related to adverse events.
“These results support the use of a 2-mg/kg Q3W dosage in patients with previously treated, advanced NSCLC,” the investigators concluded.
On Twitter@jess_craig94
A 2-mg/kg dosage of pembrolizumab given to adult patients with non–small-cell lung cancer (NSCLC) every 3 weeks is optimal to both reduce tumor size and avoid treatment-related adverse events, according to a new method of analysis.
Current methods for determining the maximum tolerated dose (MTD) based on dose-limiting toxicities may be outmoded for targeted therapies and immunotherapies, where the biologically effective dose (BED) may be much lower than the MTD.
“Using the MTD rather than the BED could expose patients to a higher dose than that necessary to achieve clinical effect and may increase toxicity, which could lower overall clinical effectiveness. Therefore, dose determination in oncology should use a multifactorial approach that includes not only clinical data from the first treatment cycle but extended clinical data, preclinical models, pharmacokinetics, pharmacodynamics, and integrated modeling and simulation,” said Dr. Manash Chatterjee of Merck, and associates (Ann Oncol. 2016 April 26. doi: 10.1093/annonc/mdw174).
Dr. Chatterjee and associates analyzed data from KEYNOTE-001 using various statistical and mathematical modeling techniques to determine the lowest effective drug dosage of pembrolizumab, a highly selective humanized IgG4 monoclonal antibody that targets the programmed death-1 receptor, for treatment of patients with NSCLC.
Patients enrolled in the KEYNOTE-001 trial received pembrolizumab at doses of 2 mg/kg every 3 weeks, 10 mg/kg every 3 weeks, or 10 mg/kg every 2 weeks until disease progression or intolerable toxicity, or until the patient or investigator decided to stop treatment. Sixty-one patients were treated with pembrolizumab at 2 mg/kg every 3 weeks, 287 patients were treated at 10 mg/kg every 3 weeks, and 202 patients were treated at 10 mg/kg every 2 weeks. PD-1 expression was assessed from biopsy samples using a prototype assay.
Decreases from baseline tumor size were observed for 67% of patients with known PD-L1 expression treated with pembrolizumab at 2 mg/kg every 3 weeks, 66% for patients treated at 10 mg/kg every 3 weeks, and 63% for patients treated at 10 mg/kg every 2 weeks.
Data on occurrence and severity of adverse events were collected throughout the study and up to 60 days after treatment was discontinued. Adverse events were graded according to the National Cancer Institute’s Common Terminology Criteria for Adverse Events (version 4.0). Treatment-related adverse events were reported for 47% of patients treated at 2 mg/kg every 3 weeks. The frequency and severity of adverse events were similar for all dosages, the investigators reported.
Exposure-efficacy was evaluated by way of exploratory regression analyses followed by nonlinear mixed effects modeling. The model found no significant difference in the exposure-efficacy relationship based on dosage. PD-L1 expression and EGFR mutation were the only two factors that explained a significant portion of interindividual variability in tumor size decrease.
Exposure-safety was evaluated via logistic regression modeling where safety was defined as the frequency of adverse events. The model revealed that there was no significant relationship between adverse events and exposure to pembrolizumab or dosage. Overall duration of treatment was the only factor that was significantly related to adverse events.
“These results support the use of a 2-mg/kg Q3W dosage in patients with previously treated, advanced NSCLC,” the investigators concluded.
On Twitter@jess_craig94
A 2-mg/kg dosage of pembrolizumab given to adult patients with non–small-cell lung cancer (NSCLC) every 3 weeks is optimal to both reduce tumor size and avoid treatment-related adverse events, according to a new method of analysis.
Current methods for determining the maximum tolerated dose (MTD) based on dose-limiting toxicities may be outmoded for targeted therapies and immunotherapies, where the biologically effective dose (BED) may be much lower than the MTD.
“Using the MTD rather than the BED could expose patients to a higher dose than that necessary to achieve clinical effect and may increase toxicity, which could lower overall clinical effectiveness. Therefore, dose determination in oncology should use a multifactorial approach that includes not only clinical data from the first treatment cycle but extended clinical data, preclinical models, pharmacokinetics, pharmacodynamics, and integrated modeling and simulation,” said Dr. Manash Chatterjee of Merck, and associates (Ann Oncol. 2016 April 26. doi: 10.1093/annonc/mdw174).
Dr. Chatterjee and associates analyzed data from KEYNOTE-001 using various statistical and mathematical modeling techniques to determine the lowest effective drug dosage of pembrolizumab, a highly selective humanized IgG4 monoclonal antibody that targets the programmed death-1 receptor, for treatment of patients with NSCLC.
Patients enrolled in the KEYNOTE-001 trial received pembrolizumab at doses of 2 mg/kg every 3 weeks, 10 mg/kg every 3 weeks, or 10 mg/kg every 2 weeks until disease progression or intolerable toxicity, or until the patient or investigator decided to stop treatment. Sixty-one patients were treated with pembrolizumab at 2 mg/kg every 3 weeks, 287 patients were treated at 10 mg/kg every 3 weeks, and 202 patients were treated at 10 mg/kg every 2 weeks. PD-1 expression was assessed from biopsy samples using a prototype assay.
Decreases from baseline tumor size were observed for 67% of patients with known PD-L1 expression treated with pembrolizumab at 2 mg/kg every 3 weeks, 66% for patients treated at 10 mg/kg every 3 weeks, and 63% for patients treated at 10 mg/kg every 2 weeks.
Data on occurrence and severity of adverse events were collected throughout the study and up to 60 days after treatment was discontinued. Adverse events were graded according to the National Cancer Institute’s Common Terminology Criteria for Adverse Events (version 4.0). Treatment-related adverse events were reported for 47% of patients treated at 2 mg/kg every 3 weeks. The frequency and severity of adverse events were similar for all dosages, the investigators reported.
Exposure-efficacy was evaluated by way of exploratory regression analyses followed by nonlinear mixed effects modeling. The model found no significant difference in the exposure-efficacy relationship based on dosage. PD-L1 expression and EGFR mutation were the only two factors that explained a significant portion of interindividual variability in tumor size decrease.
Exposure-safety was evaluated via logistic regression modeling where safety was defined as the frequency of adverse events. The model revealed that there was no significant relationship between adverse events and exposure to pembrolizumab or dosage. Overall duration of treatment was the only factor that was significantly related to adverse events.
“These results support the use of a 2-mg/kg Q3W dosage in patients with previously treated, advanced NSCLC,” the investigators concluded.
On Twitter@jess_craig94
FROM ANNALS OF ONCOLOGY
Key clinical point: A 2-mg/kg dosage of pembrolizumab given to adult NSCLC patients every 3 weeks is optimal to both reduce tumor size and avoid treatment-related adverse events.
Major finding: Mathematical models of exposure-efficacy and exposure-safety revealed no significant relationships between pembrolizumab dosage and tumor size decrease or treatment-related adverse events.
Data source: KEYNOTE-001, a multicenter, open-label, phase Ib trial of 550 patients with advanced NSCLC. The study included multiple expansion cohorts.
Disclosures: The study was supported by Merck. Six investigators reported receiving grant support, personal fees, or nonfinancial support from several companies. Thirteen investigators are full-time employees of Merck and may hold stock options in the company. The remaining four investigators reported having no financial disclosures.
CIMAvax comes to U.S. after years of development in Cuba
The Molecular Immunology Center is a campus of concrete and stone buildings with dark-tinted windows set against palm trees and pink tropical flowers. The buildings are imposing and seem to stretch endlessly along the flat terrain of Havana, Cuba. The research institute, like most buildings in Cuba, is just a few minutes’ walk to the Atlantic Ocean coastline. Inside the research and development building, investigators are working on creating 18 new cancer immunotherapy treatments from vaccines to monoclonal antibodies to synthetic cytokines. A few buildings down, in an almost 50,000–square foot manufacturing plant, a positive pressure air gradient system whirs, and stirred tank bioreactors clank in continuous motion. It is here that the CIMAvax-EGF, a therapeutic vaccine used to treat non–small-cell lung cancer (NSCLC), is manufactured, formulated, and packaged into 40-L sterile bulk bags.
The Molecular Immunology Center, also called the Center of Molecular Immunology or Centro de Immunologia Molecular, was founded in the early 1990’s and the research that eventually led to CIMAvax-EGF began almost immediately, spurred by Cuba’s high rate of lung cancer–related deaths (at the time the second leading cause of death and the leading cause of cancer mortality in the country).
The vaccine works by stimulating a patient’s own immune system to make antibodies against epidermal growth factor (EGF), which depletes circulating EGF levels and prevents EGF from binding to its receptor. “The epidermal growth factor receptor (EGFR) is a well-known oncogene. Its overactivation can induce malignant transformation of a normal cell, signaling inhibition of apoptosis, cell proliferation, angiogenesis, metastasis, and tumor-induced proinflammatory and immunosuppressive processes,” wrote Dr. Pedro Rodríguez and his associates at the Molecular Immunology Center in Cuba (MEDICC Rev. 2010;12:17-23).
“The EGFR signaling and transduction pathway can be efficiently interrupted by EGF deprivation, direct specific mAb [monoclonal antibody] receptor inhibition, or low-molecular-weight molecules competing intracellularly with adenosine triphosphate for the receptor’s tyrosine kinase activity site, with negative repercussions on cell proliferation and, consequently, on tumor development. Inducing EGF deprivation by active immunotherapy is an emerging concept developed by Cuban researchers that involves manipulating an individual’s immune response to release its own effector antibodies against EGF, thereby reducing tumor size or preventing its progression,” the investigators wrote.
Early preclinical studies for vaccine formulation began in Cuba in 1992. Over the next few years, researchers perfected the vaccine’s formula: EGF along with two immunopotentiating molecules, an adjuvant called Montanide ISA 51 and the virulent protein P64k from the microbial organism Neisseria meningitidis (Ann Oncol. 1998;9:431-5; Ann Oncol. 2003;14:461-6).
Together, the adjuvant and the virulent protein activate the immune system while the overabundance of EGF in the body directs antibodies to be made specifically against EGF and not virulent protein P64k. Although some antibodies bind the EGF introduced by the vaccine, other antibodies target cancerous cells where EGFR is overexpressed.
NSCLC was selected “because of its frequency and because EGFR is overexpressed in tissues during development and progression of lung neoplasms,” reported the investigators.
Subsequent clinical trials focused on fine-tuning administration route, dose, dosing interval, optimal combinations with other established therapies, and reducing vaccine-related side effects, which include fever, chills, nausea, headache, asthenia, and tremor.
Researchers found that administering the vaccine in “high but fractioned dose[s] in multiple anatomical sites (such as the 2 deltoid and 2 gluteal regions), thereby bringing the EGF vaccine closer to regional lymph nodes and synergizing the immune response” resulted in the best patient outcomes. In addition, significantly better patient outcomes were achieved when the vaccine was administered before and after chemotherapy.
By 1995, Cuban researchers had successfully completed five separate phase I/II clinical trials and one phase II clinical trial.
Pooling the results from the various clinical trials, non–small-cell lung cancer patients who received the vaccine and who were under the age of 60, lived an average of 4-6 months longer than patients who did not receive the vaccine or were older than 60.
By 2009 “more than 700 patients [had] received the vaccine over the years, many of them in seven clinical trials in Cuba, Canada, and the U.K.,” reported an online Cuban newspaper, Cuba Headlines.
It wasn’t until September 2011 that Cuban researchers officially began distributing the vaccine to Cuban citizens.
“The drug could turn the cancer into a manageable, chronic disease by generating antibodies against the proteins, which triggered the uncontrolled cell proliferation,” Dr. Gisela Gonzalez of the Center of Molecular Immunology said in an interview in 2009 following the official launch of the vaccine.
Today, ongoing clinical trials continue in Cuba and are also underway (at various stages) in several other countries, including the United States. The vaccine still has a long way to go before it can be hailed as a true “cancer cure,” a sentiment that many American publications expressed and one that the researchers at Roswell Park Cancer Institute who are leading the U.S. clinical trials are vehemently trying to quell. Nonetheless, the now global reach of CIMAvax may mark a promising paradigm shift in the field of oncology.
On Twitter @jess_craig94
The Molecular Immunology Center is a campus of concrete and stone buildings with dark-tinted windows set against palm trees and pink tropical flowers. The buildings are imposing and seem to stretch endlessly along the flat terrain of Havana, Cuba. The research institute, like most buildings in Cuba, is just a few minutes’ walk to the Atlantic Ocean coastline. Inside the research and development building, investigators are working on creating 18 new cancer immunotherapy treatments from vaccines to monoclonal antibodies to synthetic cytokines. A few buildings down, in an almost 50,000–square foot manufacturing plant, a positive pressure air gradient system whirs, and stirred tank bioreactors clank in continuous motion. It is here that the CIMAvax-EGF, a therapeutic vaccine used to treat non–small-cell lung cancer (NSCLC), is manufactured, formulated, and packaged into 40-L sterile bulk bags.
The Molecular Immunology Center, also called the Center of Molecular Immunology or Centro de Immunologia Molecular, was founded in the early 1990’s and the research that eventually led to CIMAvax-EGF began almost immediately, spurred by Cuba’s high rate of lung cancer–related deaths (at the time the second leading cause of death and the leading cause of cancer mortality in the country).
The vaccine works by stimulating a patient’s own immune system to make antibodies against epidermal growth factor (EGF), which depletes circulating EGF levels and prevents EGF from binding to its receptor. “The epidermal growth factor receptor (EGFR) is a well-known oncogene. Its overactivation can induce malignant transformation of a normal cell, signaling inhibition of apoptosis, cell proliferation, angiogenesis, metastasis, and tumor-induced proinflammatory and immunosuppressive processes,” wrote Dr. Pedro Rodríguez and his associates at the Molecular Immunology Center in Cuba (MEDICC Rev. 2010;12:17-23).
“The EGFR signaling and transduction pathway can be efficiently interrupted by EGF deprivation, direct specific mAb [monoclonal antibody] receptor inhibition, or low-molecular-weight molecules competing intracellularly with adenosine triphosphate for the receptor’s tyrosine kinase activity site, with negative repercussions on cell proliferation and, consequently, on tumor development. Inducing EGF deprivation by active immunotherapy is an emerging concept developed by Cuban researchers that involves manipulating an individual’s immune response to release its own effector antibodies against EGF, thereby reducing tumor size or preventing its progression,” the investigators wrote.
Early preclinical studies for vaccine formulation began in Cuba in 1992. Over the next few years, researchers perfected the vaccine’s formula: EGF along with two immunopotentiating molecules, an adjuvant called Montanide ISA 51 and the virulent protein P64k from the microbial organism Neisseria meningitidis (Ann Oncol. 1998;9:431-5; Ann Oncol. 2003;14:461-6).
Together, the adjuvant and the virulent protein activate the immune system while the overabundance of EGF in the body directs antibodies to be made specifically against EGF and not virulent protein P64k. Although some antibodies bind the EGF introduced by the vaccine, other antibodies target cancerous cells where EGFR is overexpressed.
NSCLC was selected “because of its frequency and because EGFR is overexpressed in tissues during development and progression of lung neoplasms,” reported the investigators.
Subsequent clinical trials focused on fine-tuning administration route, dose, dosing interval, optimal combinations with other established therapies, and reducing vaccine-related side effects, which include fever, chills, nausea, headache, asthenia, and tremor.
Researchers found that administering the vaccine in “high but fractioned dose[s] in multiple anatomical sites (such as the 2 deltoid and 2 gluteal regions), thereby bringing the EGF vaccine closer to regional lymph nodes and synergizing the immune response” resulted in the best patient outcomes. In addition, significantly better patient outcomes were achieved when the vaccine was administered before and after chemotherapy.
By 1995, Cuban researchers had successfully completed five separate phase I/II clinical trials and one phase II clinical trial.
Pooling the results from the various clinical trials, non–small-cell lung cancer patients who received the vaccine and who were under the age of 60, lived an average of 4-6 months longer than patients who did not receive the vaccine or were older than 60.
By 2009 “more than 700 patients [had] received the vaccine over the years, many of them in seven clinical trials in Cuba, Canada, and the U.K.,” reported an online Cuban newspaper, Cuba Headlines.
It wasn’t until September 2011 that Cuban researchers officially began distributing the vaccine to Cuban citizens.
“The drug could turn the cancer into a manageable, chronic disease by generating antibodies against the proteins, which triggered the uncontrolled cell proliferation,” Dr. Gisela Gonzalez of the Center of Molecular Immunology said in an interview in 2009 following the official launch of the vaccine.
Today, ongoing clinical trials continue in Cuba and are also underway (at various stages) in several other countries, including the United States. The vaccine still has a long way to go before it can be hailed as a true “cancer cure,” a sentiment that many American publications expressed and one that the researchers at Roswell Park Cancer Institute who are leading the U.S. clinical trials are vehemently trying to quell. Nonetheless, the now global reach of CIMAvax may mark a promising paradigm shift in the field of oncology.
On Twitter @jess_craig94
The Molecular Immunology Center is a campus of concrete and stone buildings with dark-tinted windows set against palm trees and pink tropical flowers. The buildings are imposing and seem to stretch endlessly along the flat terrain of Havana, Cuba. The research institute, like most buildings in Cuba, is just a few minutes’ walk to the Atlantic Ocean coastline. Inside the research and development building, investigators are working on creating 18 new cancer immunotherapy treatments from vaccines to monoclonal antibodies to synthetic cytokines. A few buildings down, in an almost 50,000–square foot manufacturing plant, a positive pressure air gradient system whirs, and stirred tank bioreactors clank in continuous motion. It is here that the CIMAvax-EGF, a therapeutic vaccine used to treat non–small-cell lung cancer (NSCLC), is manufactured, formulated, and packaged into 40-L sterile bulk bags.
The Molecular Immunology Center, also called the Center of Molecular Immunology or Centro de Immunologia Molecular, was founded in the early 1990’s and the research that eventually led to CIMAvax-EGF began almost immediately, spurred by Cuba’s high rate of lung cancer–related deaths (at the time the second leading cause of death and the leading cause of cancer mortality in the country).
The vaccine works by stimulating a patient’s own immune system to make antibodies against epidermal growth factor (EGF), which depletes circulating EGF levels and prevents EGF from binding to its receptor. “The epidermal growth factor receptor (EGFR) is a well-known oncogene. Its overactivation can induce malignant transformation of a normal cell, signaling inhibition of apoptosis, cell proliferation, angiogenesis, metastasis, and tumor-induced proinflammatory and immunosuppressive processes,” wrote Dr. Pedro Rodríguez and his associates at the Molecular Immunology Center in Cuba (MEDICC Rev. 2010;12:17-23).
“The EGFR signaling and transduction pathway can be efficiently interrupted by EGF deprivation, direct specific mAb [monoclonal antibody] receptor inhibition, or low-molecular-weight molecules competing intracellularly with adenosine triphosphate for the receptor’s tyrosine kinase activity site, with negative repercussions on cell proliferation and, consequently, on tumor development. Inducing EGF deprivation by active immunotherapy is an emerging concept developed by Cuban researchers that involves manipulating an individual’s immune response to release its own effector antibodies against EGF, thereby reducing tumor size or preventing its progression,” the investigators wrote.
Early preclinical studies for vaccine formulation began in Cuba in 1992. Over the next few years, researchers perfected the vaccine’s formula: EGF along with two immunopotentiating molecules, an adjuvant called Montanide ISA 51 and the virulent protein P64k from the microbial organism Neisseria meningitidis (Ann Oncol. 1998;9:431-5; Ann Oncol. 2003;14:461-6).
Together, the adjuvant and the virulent protein activate the immune system while the overabundance of EGF in the body directs antibodies to be made specifically against EGF and not virulent protein P64k. Although some antibodies bind the EGF introduced by the vaccine, other antibodies target cancerous cells where EGFR is overexpressed.
NSCLC was selected “because of its frequency and because EGFR is overexpressed in tissues during development and progression of lung neoplasms,” reported the investigators.
Subsequent clinical trials focused on fine-tuning administration route, dose, dosing interval, optimal combinations with other established therapies, and reducing vaccine-related side effects, which include fever, chills, nausea, headache, asthenia, and tremor.
Researchers found that administering the vaccine in “high but fractioned dose[s] in multiple anatomical sites (such as the 2 deltoid and 2 gluteal regions), thereby bringing the EGF vaccine closer to regional lymph nodes and synergizing the immune response” resulted in the best patient outcomes. In addition, significantly better patient outcomes were achieved when the vaccine was administered before and after chemotherapy.
By 1995, Cuban researchers had successfully completed five separate phase I/II clinical trials and one phase II clinical trial.
Pooling the results from the various clinical trials, non–small-cell lung cancer patients who received the vaccine and who were under the age of 60, lived an average of 4-6 months longer than patients who did not receive the vaccine or were older than 60.
By 2009 “more than 700 patients [had] received the vaccine over the years, many of them in seven clinical trials in Cuba, Canada, and the U.K.,” reported an online Cuban newspaper, Cuba Headlines.
It wasn’t until September 2011 that Cuban researchers officially began distributing the vaccine to Cuban citizens.
“The drug could turn the cancer into a manageable, chronic disease by generating antibodies against the proteins, which triggered the uncontrolled cell proliferation,” Dr. Gisela Gonzalez of the Center of Molecular Immunology said in an interview in 2009 following the official launch of the vaccine.
Today, ongoing clinical trials continue in Cuba and are also underway (at various stages) in several other countries, including the United States. The vaccine still has a long way to go before it can be hailed as a true “cancer cure,” a sentiment that many American publications expressed and one that the researchers at Roswell Park Cancer Institute who are leading the U.S. clinical trials are vehemently trying to quell. Nonetheless, the now global reach of CIMAvax may mark a promising paradigm shift in the field of oncology.
On Twitter @jess_craig94
Mutation pattern in non–small-cell lung cancer influenced by increased BMI, smoking
The prevalence of mutations in oncogenic driver genes is correlated to smoking dose and body mass index, according to a prospective epidemiology study of environmental factors and mutation frequencies in non–small-cell lung cancer that was published online May 9.
In the Japan Molecular Epidemiology for Lung Cancer study, Dr. Tomoya Kawaguchi and colleagues found that increased mutation frequencies in TP53, KRAS, and NFE2L2 correlated with smoking dose (P < .001 for all), whereas decreased mutation frequencies were observed in EGFR (P < .001) and CTNNB1 (P = .030). The number of KRAS mutations in smokers increased in proportion to body-mass index (BMI) increases (P = .026).
Simultaneous mutations in EGFR and CTNNB1 suggested possible biological relevance; 88% of CTNNB1 mutations (15/17) occurred with EGFR mutations. TP53 and NFE2L2 mutations were more frequent in advanced-stage disease, wrote Dr. Kawaguchi of the department of respiratory medicine at Osaka (Japan) City University and colleagues (J Clin Oncol. 2016 May 9. doi: 10.1200/JCO.2015.64.2322).
Although smoking is the most studied cause of lung cancer, about one-quarter of lung cancers worldwide occur in never-smokers.“It remains elusive which environmental factors contribute to the EGFR mutations that are frequently observed in never-smokers,” the investigators wrote. “In this study, the prevalence of EGFR mutations was higher in those who had more [environmental tobacco smoke], although this difference did not reach the level of statistical significance in the sample size. More detailed methods to detect the mutations (e.g., digital polymerase chain reaction) might yield more precise information.”
Levels of sex hormones were not significant factors in mutation frequencies, but the investigators found that estrogen receptor was more highly expressed in never-smokers than smokers, and the presence of estrogen receptor was associated with EGFR mutations in younger patients.
The investigators studied environmental influences on lung cancer by collecting information by questionnaire and by detecting mutations in 72 candidate genes from 876 patients with stage I to IIIB non–small-cell lung cancer (441 ever- and 435 never-smokers). In total, 622 patients had at least one mutation, and 860 mutations were observed. Dr. Kawaguchi and colleagues also examined patterns of estrogen-receptor expression by immunohistochemical staining and evidence of human papillomavirus (HPV) infection by a polymerase chain reaction–based microarray system.
Contrary to retrospective analyses that had pointed to a link between HPV and NSCLC, this prospective study showed little evidence for HPV in early NSCLC.
Dr. Kawaguchi reported having financial ties to Chugai Pharmaceutical and Eli Lilly. Several coauthors reported tis to industry sources.
The prevalence of mutations in oncogenic driver genes is correlated to smoking dose and body mass index, according to a prospective epidemiology study of environmental factors and mutation frequencies in non–small-cell lung cancer that was published online May 9.
In the Japan Molecular Epidemiology for Lung Cancer study, Dr. Tomoya Kawaguchi and colleagues found that increased mutation frequencies in TP53, KRAS, and NFE2L2 correlated with smoking dose (P < .001 for all), whereas decreased mutation frequencies were observed in EGFR (P < .001) and CTNNB1 (P = .030). The number of KRAS mutations in smokers increased in proportion to body-mass index (BMI) increases (P = .026).
Simultaneous mutations in EGFR and CTNNB1 suggested possible biological relevance; 88% of CTNNB1 mutations (15/17) occurred with EGFR mutations. TP53 and NFE2L2 mutations were more frequent in advanced-stage disease, wrote Dr. Kawaguchi of the department of respiratory medicine at Osaka (Japan) City University and colleagues (J Clin Oncol. 2016 May 9. doi: 10.1200/JCO.2015.64.2322).
Although smoking is the most studied cause of lung cancer, about one-quarter of lung cancers worldwide occur in never-smokers.“It remains elusive which environmental factors contribute to the EGFR mutations that are frequently observed in never-smokers,” the investigators wrote. “In this study, the prevalence of EGFR mutations was higher in those who had more [environmental tobacco smoke], although this difference did not reach the level of statistical significance in the sample size. More detailed methods to detect the mutations (e.g., digital polymerase chain reaction) might yield more precise information.”
Levels of sex hormones were not significant factors in mutation frequencies, but the investigators found that estrogen receptor was more highly expressed in never-smokers than smokers, and the presence of estrogen receptor was associated with EGFR mutations in younger patients.
The investigators studied environmental influences on lung cancer by collecting information by questionnaire and by detecting mutations in 72 candidate genes from 876 patients with stage I to IIIB non–small-cell lung cancer (441 ever- and 435 never-smokers). In total, 622 patients had at least one mutation, and 860 mutations were observed. Dr. Kawaguchi and colleagues also examined patterns of estrogen-receptor expression by immunohistochemical staining and evidence of human papillomavirus (HPV) infection by a polymerase chain reaction–based microarray system.
Contrary to retrospective analyses that had pointed to a link between HPV and NSCLC, this prospective study showed little evidence for HPV in early NSCLC.
Dr. Kawaguchi reported having financial ties to Chugai Pharmaceutical and Eli Lilly. Several coauthors reported tis to industry sources.
The prevalence of mutations in oncogenic driver genes is correlated to smoking dose and body mass index, according to a prospective epidemiology study of environmental factors and mutation frequencies in non–small-cell lung cancer that was published online May 9.
In the Japan Molecular Epidemiology for Lung Cancer study, Dr. Tomoya Kawaguchi and colleagues found that increased mutation frequencies in TP53, KRAS, and NFE2L2 correlated with smoking dose (P < .001 for all), whereas decreased mutation frequencies were observed in EGFR (P < .001) and CTNNB1 (P = .030). The number of KRAS mutations in smokers increased in proportion to body-mass index (BMI) increases (P = .026).
Simultaneous mutations in EGFR and CTNNB1 suggested possible biological relevance; 88% of CTNNB1 mutations (15/17) occurred with EGFR mutations. TP53 and NFE2L2 mutations were more frequent in advanced-stage disease, wrote Dr. Kawaguchi of the department of respiratory medicine at Osaka (Japan) City University and colleagues (J Clin Oncol. 2016 May 9. doi: 10.1200/JCO.2015.64.2322).
Although smoking is the most studied cause of lung cancer, about one-quarter of lung cancers worldwide occur in never-smokers.“It remains elusive which environmental factors contribute to the EGFR mutations that are frequently observed in never-smokers,” the investigators wrote. “In this study, the prevalence of EGFR mutations was higher in those who had more [environmental tobacco smoke], although this difference did not reach the level of statistical significance in the sample size. More detailed methods to detect the mutations (e.g., digital polymerase chain reaction) might yield more precise information.”
Levels of sex hormones were not significant factors in mutation frequencies, but the investigators found that estrogen receptor was more highly expressed in never-smokers than smokers, and the presence of estrogen receptor was associated with EGFR mutations in younger patients.
The investigators studied environmental influences on lung cancer by collecting information by questionnaire and by detecting mutations in 72 candidate genes from 876 patients with stage I to IIIB non–small-cell lung cancer (441 ever- and 435 never-smokers). In total, 622 patients had at least one mutation, and 860 mutations were observed. Dr. Kawaguchi and colleagues also examined patterns of estrogen-receptor expression by immunohistochemical staining and evidence of human papillomavirus (HPV) infection by a polymerase chain reaction–based microarray system.
Contrary to retrospective analyses that had pointed to a link between HPV and NSCLC, this prospective study showed little evidence for HPV in early NSCLC.
Dr. Kawaguchi reported having financial ties to Chugai Pharmaceutical and Eli Lilly. Several coauthors reported tis to industry sources.
FROM THE JOURNAL OF CLINICAL ONCOLOGY
Key clinical point: A prospective epidemiology study found that smoking dose and body-mass index correlated with mutation patterns in non–small-cell lung cancer (NSCLC).
Major finding: The prevalence of TP53, KRAS, and NFE2L2 increased proportionally with smoking dose (P < .001 for all), whereas mutation prevalence in EGFR (P < .001) and CTNNB1 (P = .030) decreased; KRAS mutations were observed more frequently in proportion to increasing BMI in ever-smokers.
Data source: The Japan Molecular Epidemiology for Lung Cancer Study examined mutations in 876 patients with stage I to IIIB NSCLC.
Disclosures: Dr. Kawaguchi reported having financial ties to Chugai Pharmaceutical and Eli Lilly. Several coauthors reported ties to industry sources.


