Hypogonadism after testicular cancer treatment can have lifelong impact

Article Type
Changed
Fri, 01/18/2019 - 16:50

 

– Hypogonadism may compromise the long-term health outlook for many younger men who have been successfully treated for testicular cancer, according to an analysis conducted by the Platinum Study Group.

“Today, 95% of all testicular cancer patients are cured of their disease thanks to cisplatin-based chemotherapy. Nowadays, testicular cancer survivors can expect to live for over 40 years from the time of their diagnosis,” lead investigator Mohammad Issam Abu Zaid, MBBS, said in a press briefing at the annual meeting of the American Society of Clinical Oncology. “However, they are at risk of other health problems that may be related to their cancer treatment, including late complications from chemotherapy.”

Dr. Abu Zaid, assistant professor of medicine at Indiana University, Indianapolis
Susan London/Frontline Medical News
Dr. Abu Zaid
The analysis of almost 500 younger testicular cancer survivors who had received cisplatin-based chemotherapy found that nearly 4 in 10 had hypogonadism. Compared with unaffected peers, men in this group were up to four times more likely to be taking medication for a variety of chronic conditions: dyslipidemia, hypertension, diabetes, erectile dysfunction, and anxiety or depression.

“Testicular cancer survivors, especially those treated with chemotherapy, are at increased risk for hypogonadism, a problem that can be associated with predisposing factors for heart disease,” summarized Dr. Abu Zaid, of Indiana University in Indianapolis. “Mitigating approaches are the usual weight control, exercise, and monitoring of blood pressure and cholesterol levels.”

Dr. Timothy D. Gilligan, a medical oncologist at the Cleveland Clinic
Susan London/Frontline Medical News
Dr. Timothy D. Gilligan
“We will never omit cisplatin from the treatment regimen to prevent complications, but recognizing and treating symptomatic hypogonadism can improve quality of life and lessen adverse health outcomes, especially those related to the metabolic syndrome and resulting in diabetes, hyperlipidemia, and early cardiac problems,” he asserted.
 

Expert perspective

“This is an important study, and it sends a loud message to those of us who take care of testis cancer patients, my area of expertise. ... We need to watch for hypogonadism, and we need to ask survivors about it. We need to examine them thinking about it, and, in patients who we are worried [they] might have hypogonadism, we need to do blood tests for testosterone and other hormone levels,” commented ASCO Expert Timothy D. Gilligan, MD, MSc, of the Cleveland Clinic in Ohio. “These are young patients, they have many years of life, so it’s many decades of suffering from consequences of this if it’s undetected.”

The findings were not surprising based on his personal experience and on evidence from the prostate cancer field showing the adverse metabolic effects of withdrawing testosterone, he said. Although the prevalence of hypogonadism found in the study was higher than that found in other studies, given the large size of the cohort, it should be taken seriously. Additionally, even if the true prevalence is somewhat lower, the absolute number of survivors affected would be substantial.

“We need to be cautious though and make sure people don’t misunderstand and think that this means we should test testosterone levels in all patients, which is a risky thing to do because the definition of normal testosterone is very fuzzy,” Dr. Gilligan stressed. “There is a wide range of normal, and what’s normal for me may not be the same as what’s normal for another man. So, looking for symptoms is really what guides this work, and, when there are symptoms, then testing is important.”

Study details

The Platinum Study is a large, ongoing, multicenter North American–based cohort study of testicular cancer survivors who received cisplatin as part of their chemotherapy.

Dr. Abu Zaid and his colleagues analyzed data from 491 participants who were aged 50 years or younger at diagnosis. All completed questionnaires addressing comorbidities, medications, and health behaviors; underwent physical examination; had measurement of testosterone levels; and had a genetic analysis.

The men were by and large young at the time of evaluation, with a mean age of just 38 years, he reported in the press briefing and a poster session at the meeting.

Overall, 38.5% had hypogonadism, defined in the study as a serum testosterone level of 3 ng/mL or lower or as use of testosterone replacement therapy.

In a multivariate analysis, survivors’ odds of hypogonadism increased with age (odds ratio, 1.41 per 10-year increment; P = .007) and were higher for those having a body mass index of 25 kg/m2 or greater, vs. lower (OR, 2.22; P = .003). On the other hand, there was a trend whereby survivors who engaged in any vigorous-intensity physical activity, vs. none, were less likely to have hypogonadism (OR, 0.64; P = .06).

The odds rose with number of risk alleles in the sex hormone–binding globulin gene, but findings were not significant, Dr. Abu Zaid said. They were also statistically indistinguishable across groups differing with respect to the chemotherapy regimen received and socioeconomic factors.

Compared with counterparts having normal testosterone levels, survivors having hypogonadism were up to four times more likely to be taking medication to treat dyslipidemia (20.1% vs. 6.0%; P less than .001), hypertension (18.5% vs. 10.6%; P = .013), erectile dysfunction (19.6% vs. 11.9%; P = .018), diabetes (5.8% vs. 2.6%; P = .067), and anxiety or depression (14.8% vs. 9.3%; P = .060).

Testicular cancer survivors should not universally have testosterone testing, agreed Dr. Abu Zaid. Such a practice might lead to overtreatment, and, in older men at least, use of testosterone itself has been linked to elevated cardiovascular risk.

“You really want to treat somebody who has symptoms related to hypogonadism: constant fatigue, night sweats, and depressed mood and some other symptoms that are well known to the clinician,” he maintained. “At the moment, we have no studies done looking at testosterone replacement in young men. I would hypothesize that we actually help them by giving them testosterone, and that’s what I tell my patients.”

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

 

– Hypogonadism may compromise the long-term health outlook for many younger men who have been successfully treated for testicular cancer, according to an analysis conducted by the Platinum Study Group.

“Today, 95% of all testicular cancer patients are cured of their disease thanks to cisplatin-based chemotherapy. Nowadays, testicular cancer survivors can expect to live for over 40 years from the time of their diagnosis,” lead investigator Mohammad Issam Abu Zaid, MBBS, said in a press briefing at the annual meeting of the American Society of Clinical Oncology. “However, they are at risk of other health problems that may be related to their cancer treatment, including late complications from chemotherapy.”

Dr. Abu Zaid, assistant professor of medicine at Indiana University, Indianapolis
Susan London/Frontline Medical News
Dr. Abu Zaid
The analysis of almost 500 younger testicular cancer survivors who had received cisplatin-based chemotherapy found that nearly 4 in 10 had hypogonadism. Compared with unaffected peers, men in this group were up to four times more likely to be taking medication for a variety of chronic conditions: dyslipidemia, hypertension, diabetes, erectile dysfunction, and anxiety or depression.

“Testicular cancer survivors, especially those treated with chemotherapy, are at increased risk for hypogonadism, a problem that can be associated with predisposing factors for heart disease,” summarized Dr. Abu Zaid, of Indiana University in Indianapolis. “Mitigating approaches are the usual weight control, exercise, and monitoring of blood pressure and cholesterol levels.”

Dr. Timothy D. Gilligan, a medical oncologist at the Cleveland Clinic
Susan London/Frontline Medical News
Dr. Timothy D. Gilligan
“We will never omit cisplatin from the treatment regimen to prevent complications, but recognizing and treating symptomatic hypogonadism can improve quality of life and lessen adverse health outcomes, especially those related to the metabolic syndrome and resulting in diabetes, hyperlipidemia, and early cardiac problems,” he asserted.
 

Expert perspective

“This is an important study, and it sends a loud message to those of us who take care of testis cancer patients, my area of expertise. ... We need to watch for hypogonadism, and we need to ask survivors about it. We need to examine them thinking about it, and, in patients who we are worried [they] might have hypogonadism, we need to do blood tests for testosterone and other hormone levels,” commented ASCO Expert Timothy D. Gilligan, MD, MSc, of the Cleveland Clinic in Ohio. “These are young patients, they have many years of life, so it’s many decades of suffering from consequences of this if it’s undetected.”

The findings were not surprising based on his personal experience and on evidence from the prostate cancer field showing the adverse metabolic effects of withdrawing testosterone, he said. Although the prevalence of hypogonadism found in the study was higher than that found in other studies, given the large size of the cohort, it should be taken seriously. Additionally, even if the true prevalence is somewhat lower, the absolute number of survivors affected would be substantial.

“We need to be cautious though and make sure people don’t misunderstand and think that this means we should test testosterone levels in all patients, which is a risky thing to do because the definition of normal testosterone is very fuzzy,” Dr. Gilligan stressed. “There is a wide range of normal, and what’s normal for me may not be the same as what’s normal for another man. So, looking for symptoms is really what guides this work, and, when there are symptoms, then testing is important.”

Study details

The Platinum Study is a large, ongoing, multicenter North American–based cohort study of testicular cancer survivors who received cisplatin as part of their chemotherapy.

Dr. Abu Zaid and his colleagues analyzed data from 491 participants who were aged 50 years or younger at diagnosis. All completed questionnaires addressing comorbidities, medications, and health behaviors; underwent physical examination; had measurement of testosterone levels; and had a genetic analysis.

The men were by and large young at the time of evaluation, with a mean age of just 38 years, he reported in the press briefing and a poster session at the meeting.

Overall, 38.5% had hypogonadism, defined in the study as a serum testosterone level of 3 ng/mL or lower or as use of testosterone replacement therapy.

In a multivariate analysis, survivors’ odds of hypogonadism increased with age (odds ratio, 1.41 per 10-year increment; P = .007) and were higher for those having a body mass index of 25 kg/m2 or greater, vs. lower (OR, 2.22; P = .003). On the other hand, there was a trend whereby survivors who engaged in any vigorous-intensity physical activity, vs. none, were less likely to have hypogonadism (OR, 0.64; P = .06).

The odds rose with number of risk alleles in the sex hormone–binding globulin gene, but findings were not significant, Dr. Abu Zaid said. They were also statistically indistinguishable across groups differing with respect to the chemotherapy regimen received and socioeconomic factors.

Compared with counterparts having normal testosterone levels, survivors having hypogonadism were up to four times more likely to be taking medication to treat dyslipidemia (20.1% vs. 6.0%; P less than .001), hypertension (18.5% vs. 10.6%; P = .013), erectile dysfunction (19.6% vs. 11.9%; P = .018), diabetes (5.8% vs. 2.6%; P = .067), and anxiety or depression (14.8% vs. 9.3%; P = .060).

Testicular cancer survivors should not universally have testosterone testing, agreed Dr. Abu Zaid. Such a practice might lead to overtreatment, and, in older men at least, use of testosterone itself has been linked to elevated cardiovascular risk.

“You really want to treat somebody who has symptoms related to hypogonadism: constant fatigue, night sweats, and depressed mood and some other symptoms that are well known to the clinician,” he maintained. “At the moment, we have no studies done looking at testosterone replacement in young men. I would hypothesize that we actually help them by giving them testosterone, and that’s what I tell my patients.”

 

– Hypogonadism may compromise the long-term health outlook for many younger men who have been successfully treated for testicular cancer, according to an analysis conducted by the Platinum Study Group.

“Today, 95% of all testicular cancer patients are cured of their disease thanks to cisplatin-based chemotherapy. Nowadays, testicular cancer survivors can expect to live for over 40 years from the time of their diagnosis,” lead investigator Mohammad Issam Abu Zaid, MBBS, said in a press briefing at the annual meeting of the American Society of Clinical Oncology. “However, they are at risk of other health problems that may be related to their cancer treatment, including late complications from chemotherapy.”

Dr. Abu Zaid, assistant professor of medicine at Indiana University, Indianapolis
Susan London/Frontline Medical News
Dr. Abu Zaid
The analysis of almost 500 younger testicular cancer survivors who had received cisplatin-based chemotherapy found that nearly 4 in 10 had hypogonadism. Compared with unaffected peers, men in this group were up to four times more likely to be taking medication for a variety of chronic conditions: dyslipidemia, hypertension, diabetes, erectile dysfunction, and anxiety or depression.

“Testicular cancer survivors, especially those treated with chemotherapy, are at increased risk for hypogonadism, a problem that can be associated with predisposing factors for heart disease,” summarized Dr. Abu Zaid, of Indiana University in Indianapolis. “Mitigating approaches are the usual weight control, exercise, and monitoring of blood pressure and cholesterol levels.”

Dr. Timothy D. Gilligan, a medical oncologist at the Cleveland Clinic
Susan London/Frontline Medical News
Dr. Timothy D. Gilligan
“We will never omit cisplatin from the treatment regimen to prevent complications, but recognizing and treating symptomatic hypogonadism can improve quality of life and lessen adverse health outcomes, especially those related to the metabolic syndrome and resulting in diabetes, hyperlipidemia, and early cardiac problems,” he asserted.
 

Expert perspective

“This is an important study, and it sends a loud message to those of us who take care of testis cancer patients, my area of expertise. ... We need to watch for hypogonadism, and we need to ask survivors about it. We need to examine them thinking about it, and, in patients who we are worried [they] might have hypogonadism, we need to do blood tests for testosterone and other hormone levels,” commented ASCO Expert Timothy D. Gilligan, MD, MSc, of the Cleveland Clinic in Ohio. “These are young patients, they have many years of life, so it’s many decades of suffering from consequences of this if it’s undetected.”

The findings were not surprising based on his personal experience and on evidence from the prostate cancer field showing the adverse metabolic effects of withdrawing testosterone, he said. Although the prevalence of hypogonadism found in the study was higher than that found in other studies, given the large size of the cohort, it should be taken seriously. Additionally, even if the true prevalence is somewhat lower, the absolute number of survivors affected would be substantial.

“We need to be cautious though and make sure people don’t misunderstand and think that this means we should test testosterone levels in all patients, which is a risky thing to do because the definition of normal testosterone is very fuzzy,” Dr. Gilligan stressed. “There is a wide range of normal, and what’s normal for me may not be the same as what’s normal for another man. So, looking for symptoms is really what guides this work, and, when there are symptoms, then testing is important.”

Study details

The Platinum Study is a large, ongoing, multicenter North American–based cohort study of testicular cancer survivors who received cisplatin as part of their chemotherapy.

Dr. Abu Zaid and his colleagues analyzed data from 491 participants who were aged 50 years or younger at diagnosis. All completed questionnaires addressing comorbidities, medications, and health behaviors; underwent physical examination; had measurement of testosterone levels; and had a genetic analysis.

The men were by and large young at the time of evaluation, with a mean age of just 38 years, he reported in the press briefing and a poster session at the meeting.

Overall, 38.5% had hypogonadism, defined in the study as a serum testosterone level of 3 ng/mL or lower or as use of testosterone replacement therapy.

In a multivariate analysis, survivors’ odds of hypogonadism increased with age (odds ratio, 1.41 per 10-year increment; P = .007) and were higher for those having a body mass index of 25 kg/m2 or greater, vs. lower (OR, 2.22; P = .003). On the other hand, there was a trend whereby survivors who engaged in any vigorous-intensity physical activity, vs. none, were less likely to have hypogonadism (OR, 0.64; P = .06).

The odds rose with number of risk alleles in the sex hormone–binding globulin gene, but findings were not significant, Dr. Abu Zaid said. They were also statistically indistinguishable across groups differing with respect to the chemotherapy regimen received and socioeconomic factors.

Compared with counterparts having normal testosterone levels, survivors having hypogonadism were up to four times more likely to be taking medication to treat dyslipidemia (20.1% vs. 6.0%; P less than .001), hypertension (18.5% vs. 10.6%; P = .013), erectile dysfunction (19.6% vs. 11.9%; P = .018), diabetes (5.8% vs. 2.6%; P = .067), and anxiety or depression (14.8% vs. 9.3%; P = .060).

Testicular cancer survivors should not universally have testosterone testing, agreed Dr. Abu Zaid. Such a practice might lead to overtreatment, and, in older men at least, use of testosterone itself has been linked to elevated cardiovascular risk.

“You really want to treat somebody who has symptoms related to hypogonadism: constant fatigue, night sweats, and depressed mood and some other symptoms that are well known to the clinician,” he maintained. “At the moment, we have no studies done looking at testosterone replacement in young men. I would hypothesize that we actually help them by giving them testosterone, and that’s what I tell my patients.”

Publications
Publications
Topics
Article Type
Sections
Article Source

AT ASCO 2017

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

 

Key clinical point: Hypogonadism is prevalent among young testicular cancer survivors treated with cisplatin and confers elevated risks of chronic conditions.

Major finding: Fully 38.5% of survivors had low testosterone levels, and men in this group were more likely to be taking medications for dyslipidemia, hypertension, diabetes, erectile dysfunction, and anxiety or depression.

Data source: A cohort study of 491 testicular cancer survivors who had been treated with cisplatin-based chemotherapy (Platinum Study).

Disclosures: Dr. Abu Zaid reported that he had no relevant disclosures.

VIDEO: Metastatic Trial Search links MBC patients to relevant trials

Article Type
Changed
Wed, 01/04/2023 - 16:59


CHICAGO– Metastatic Trial Search was launched in 2015 by ClinicalTrials.org to make it easier for patients with metastatic breast cancer to consider clinical trials as a routine option as they are making treatment decisions with their physicians.

In a video interview, Shirley A. Mertz, JD, president of the Metastatic Breast Cancer Network, describes the tool, the mixed response from physicians, the barriers to trial participation still faced by patients, and the tweaked version 2.0 of the search tool, expected to launch by the end of this year.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event


CHICAGO– Metastatic Trial Search was launched in 2015 by ClinicalTrials.org to make it easier for patients with metastatic breast cancer to consider clinical trials as a routine option as they are making treatment decisions with their physicians.

In a video interview, Shirley A. Mertz, JD, president of the Metastatic Breast Cancer Network, describes the tool, the mixed response from physicians, the barriers to trial participation still faced by patients, and the tweaked version 2.0 of the search tool, expected to launch by the end of this year.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel


CHICAGO– Metastatic Trial Search was launched in 2015 by ClinicalTrials.org to make it easier for patients with metastatic breast cancer to consider clinical trials as a routine option as they are making treatment decisions with their physicians.

In a video interview, Shirley A. Mertz, JD, president of the Metastatic Breast Cancer Network, describes the tool, the mixed response from physicians, the barriers to trial participation still faced by patients, and the tweaked version 2.0 of the search tool, expected to launch by the end of this year.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Publications
Publications
Topics
Article Type
Sections
Article Source

AT ASCO 2017

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

Combo with daratumumab could be alternative to ASCT in MM

Article Type
Changed
Tue, 06/13/2017 - 01:00
Display Headline
Combo with daratumumab could be alternative to ASCT in MM

Photo © ASCO/Scott Morgan 2017
Attendees at ASCO 2017 Annual Meeting

CHICAGO—Results of an open-label phase 1b study of daratumumab combined with carfilzomib, lenalidomide, and dexamethasone (KRd) in newly diagnosed multiple myeloma (MM) patients have shown the combination to be highly effective, with an overall response rate of 100%.

Ninety-one percent of patients achieved a very good partial response (VGPR) or better, and 43% achieved a complete response (CR) or better.

Investigators had hypothesized that rather than using autologous stem cell transplant (ASCT) to improve results of treatment with KRd, the combination could alternatively be improved by incorporating daratumumab into a KRd regimen.

Andrzej Jakubowiak, MD, of the University of Chicago Medical Center in Illinois, presented the findings of the MMY1001 study at the 2017 ASCO Annual Meeting (abstract 8000*).

“I think what was one of the more important developments in myeloma last year,” Dr Jakubowiak said, “was data from randomized studies showing that adding daratumumab to either lenalidomide and dexamethasone in the POLLUX study or bortezomib and dexamethasone, a proteasome inhibitor, in the CASTOR study, improves responses, depth of response, and . . .  dramatically improved progression-free survival.”

“[W]e have now the rationale to potentially combine daratumumab with both an IMiD and proteasome inhibitor,” he explained, “which led to the development of this phase 1b study in which we combined daratumumab with KRd and evaluated tolerability and efficacy.”

Study design

Twenty-two transplant-eligible or -ineligible newly diagnosed MM patients were enrolled on the study.

Treatment duration was planned to be 13 cycles or less and patients had the option to move to transplant after 4 cycles.

They could have no clinically significant cardiac disease and echocardiogram was required prior to transplant.

The dosing schedule was the established dosing schema for daratumumab and KRd with 2 notable differences in the 28-day cycles.

First, the daratumumab dose was a split dose. So patients received 8 mg/kg on days 1-2 of cycle 1, 16 mg/kg a week on cycle 2, 16 mg/kg every 2 weeks on cycles 3 – 6, and every 4th week thereafter.

The second difference was carfilzomib dosing was a weekly regimen with escalation from 20 mg/m2 on day 1, cycle 1 to 70 mg/m2 on day 8 of cycle 1.

Lenalidomide (25 mg on days 1-21 of each cycle) and dexamethasone (40 mg/week) were the standard regimens for these drugs.

The primary endpoint was safety and tolerability. The secondary endpoint was overall response rate (ORR), duration of response, time to response, and infusion-related reactions (IRR).

The study also had an exploratory endpoint of progression-free survival (PFS).

Baseline characteristics

Patients were a median age of 59.5 years (range 34 – 74). About two thirds were younger than 65 and one third were between 65 and 75.

A little over half were male and most (86%) were white.

A little more than half (55%) had an ECOG score of 0, 41% were ECOG 1, and 5% were ECOG 2.

Patient disposition

As of the cutoff date of March 24, 8 of the 22 patients enrolled (36%) discontinued treatment: 1 due to an adverse event (AE), 1 due to progressive disease, and 6 patients (27%) proceeded to ASCT.

Dr Jakubowiak pointed out that response was censored at this point for patients who proceeded to transplant.

The median follow-up was 10.8 months (range, 4.0 – 12.5) and the median number of treatment cycles was 11.5 (range, 1.0 – 13.0).

“What is of interest to many of us,” Dr Jakubowiak said, “is that patients were escalated to the planned dose of 70 mg/m2 by cycle 2 except for 3 patients.”

 

 

Of the 3, 1 discontinued before day 1 of the second cycle due to toxicity, 1 had a dose reduction to 56 mg/m2 at day of the second cycle, and 1 escalated to 70 mg/m2 at day 8 of cycle 3.

Ultimately, all patients who remained on study were able to escalate to 70 mg/m2.

Safety

The hematologic treatment-emergent adverse events (TEAE) generally followed what has been observed in similar studies before, Dr Jakubowiak noted.

Hematologic TEAEs of all grades occurring in 30% or more of patients were lymphopenia (68%), thrombocytopenia (55%), anemia (46%), leukopenia (41%), and neutropenia (32%).

The most common non-hematologic TEAEs of all grades occurring in 30% of patients or more were diarrhea (73%), upper respiratory infection (59%) cough, constipation, and fatigue (50% each), dyspnea and insomnia (46%), nausea, rash, and back pain (41%), muscle spasm (36%), and vomiting, pain in extremity, hyperglycemia, and increased ALT (32%).

The most common grade 3/4 TEAEs were infrequent and many events had none of grade 3/4 severity.

The safety profile is consistent with what was previously reported for daratumumab or KRd, Dr Jakubowiak affirmed.

Serious TEAEs

Serious TEAEs occurred in 10 patients (46%), with many occurring in just 1 patient. Pulmonary embolism (PE) was the most frequent, occurring in 3 patients.

All patients were required to be on aspirin prophylaxis and 1 of the patients who had a PE discontinued therapy.

The number of patients with a serious TEAE reasonably related to an individual study drug were 3 (14%) for daratumumab, 5 (23%) for carfilzomib, 5 (23%) for lenalidomide, and 2 (9%) for dexamethasone.

The TEAEs of interest—tachycardia, congestive heart failure, and hypertension—occurred in a single patient each.

Overall, serious TEAEs were consistent with previous reports from KRd studies.

Echocardiogram assessment

Investigators conducted 30 systemic evaluations on the impact of this regimen on heart function.  The investigators observed no change from baseline through the duration of treatment in patients’ left ventricular ejection fractions.

One patient developed congestive heart failure, possibly related to daratumumab or carfilzomib. This patient resumed treatment with a reduced carfilzomib dose, elected ASCT on study day 113, and ended treatment with a VGPR.

“In all,” Dr Jakubowiak said, “we feel that there is no apparent signal of adverse impact of the addition of daratumumab on cardiac function.”

Infusion times and reactions

Overall, IRRs occurred in 27% of the patients, “which appears lower than with previous daratumumab studies,” Dr Jakubowiak noted. And IRRs occurred more frequently during the first infusion than subsequent infusions.

The split-dose infusion time was very similar to that of second and subsequent cycles.

There were limited events related to infusions. All were grade 1 or 2 and most occurred in only a single patient.

Response rate

The median number of treatment cycles administered was 11.5 (range, 2.0 – 13.0). The best response was 100% PR or better, 91% achieved VGPR or better, 42% CR or better, and 29% a stringent CR.

The depth of response improved with duration of treatment. For example, the sCR rate increased from 5% after 4 cycles to 29% at the end of treatment.

PFS was an exploratory endpoint. One patient progressed at 10.8 months and the 12-month PFS rate was 94% with all patients alive.

Stem cell harvest and ASCT

“For many of us,” Dr Jakubowiak commented, “it’s also of interest how this regimen will impact stem cell harvest.”

Nineteen of 22 patients were deemed to be transplant eligible, and the median number of CD34+ cells collected from them was 10.4 x 10cells/kg.

 

 

Patients had a median of 5 treatment cycles prior to stem cell harvest, and 14 (74%) had a VGPR or better prior to harvest.

The investigators believe stem cell yield was consistent with previous KRd studies.

Dr Jakubowiak commented that the deepening of response over time “is a phenomenon we think is important. . . . In all, the data from this small phase 1b study provide support for further evaluation of this regimen in newly diagnosed myeloma."

The study was funded by Janssen Research and Development, LLC. 

*Data presented during the meeting differ from the abstract.

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

Photo © ASCO/Scott Morgan 2017
Attendees at ASCO 2017 Annual Meeting

CHICAGO—Results of an open-label phase 1b study of daratumumab combined with carfilzomib, lenalidomide, and dexamethasone (KRd) in newly diagnosed multiple myeloma (MM) patients have shown the combination to be highly effective, with an overall response rate of 100%.

Ninety-one percent of patients achieved a very good partial response (VGPR) or better, and 43% achieved a complete response (CR) or better.

Investigators had hypothesized that rather than using autologous stem cell transplant (ASCT) to improve results of treatment with KRd, the combination could alternatively be improved by incorporating daratumumab into a KRd regimen.

Andrzej Jakubowiak, MD, of the University of Chicago Medical Center in Illinois, presented the findings of the MMY1001 study at the 2017 ASCO Annual Meeting (abstract 8000*).

“I think what was one of the more important developments in myeloma last year,” Dr Jakubowiak said, “was data from randomized studies showing that adding daratumumab to either lenalidomide and dexamethasone in the POLLUX study or bortezomib and dexamethasone, a proteasome inhibitor, in the CASTOR study, improves responses, depth of response, and . . .  dramatically improved progression-free survival.”

“[W]e have now the rationale to potentially combine daratumumab with both an IMiD and proteasome inhibitor,” he explained, “which led to the development of this phase 1b study in which we combined daratumumab with KRd and evaluated tolerability and efficacy.”

Study design

Twenty-two transplant-eligible or -ineligible newly diagnosed MM patients were enrolled on the study.

Treatment duration was planned to be 13 cycles or less and patients had the option to move to transplant after 4 cycles.

They could have no clinically significant cardiac disease and echocardiogram was required prior to transplant.

The dosing schedule was the established dosing schema for daratumumab and KRd with 2 notable differences in the 28-day cycles.

First, the daratumumab dose was a split dose. So patients received 8 mg/kg on days 1-2 of cycle 1, 16 mg/kg a week on cycle 2, 16 mg/kg every 2 weeks on cycles 3 – 6, and every 4th week thereafter.

The second difference was carfilzomib dosing was a weekly regimen with escalation from 20 mg/m2 on day 1, cycle 1 to 70 mg/m2 on day 8 of cycle 1.

Lenalidomide (25 mg on days 1-21 of each cycle) and dexamethasone (40 mg/week) were the standard regimens for these drugs.

The primary endpoint was safety and tolerability. The secondary endpoint was overall response rate (ORR), duration of response, time to response, and infusion-related reactions (IRR).

The study also had an exploratory endpoint of progression-free survival (PFS).

Baseline characteristics

Patients were a median age of 59.5 years (range 34 – 74). About two thirds were younger than 65 and one third were between 65 and 75.

A little over half were male and most (86%) were white.

A little more than half (55%) had an ECOG score of 0, 41% were ECOG 1, and 5% were ECOG 2.

Patient disposition

As of the cutoff date of March 24, 8 of the 22 patients enrolled (36%) discontinued treatment: 1 due to an adverse event (AE), 1 due to progressive disease, and 6 patients (27%) proceeded to ASCT.

Dr Jakubowiak pointed out that response was censored at this point for patients who proceeded to transplant.

The median follow-up was 10.8 months (range, 4.0 – 12.5) and the median number of treatment cycles was 11.5 (range, 1.0 – 13.0).

“What is of interest to many of us,” Dr Jakubowiak said, “is that patients were escalated to the planned dose of 70 mg/m2 by cycle 2 except for 3 patients.”

 

 

Of the 3, 1 discontinued before day 1 of the second cycle due to toxicity, 1 had a dose reduction to 56 mg/m2 at day of the second cycle, and 1 escalated to 70 mg/m2 at day 8 of cycle 3.

Ultimately, all patients who remained on study were able to escalate to 70 mg/m2.

Safety

The hematologic treatment-emergent adverse events (TEAE) generally followed what has been observed in similar studies before, Dr Jakubowiak noted.

Hematologic TEAEs of all grades occurring in 30% or more of patients were lymphopenia (68%), thrombocytopenia (55%), anemia (46%), leukopenia (41%), and neutropenia (32%).

The most common non-hematologic TEAEs of all grades occurring in 30% of patients or more were diarrhea (73%), upper respiratory infection (59%) cough, constipation, and fatigue (50% each), dyspnea and insomnia (46%), nausea, rash, and back pain (41%), muscle spasm (36%), and vomiting, pain in extremity, hyperglycemia, and increased ALT (32%).

The most common grade 3/4 TEAEs were infrequent and many events had none of grade 3/4 severity.

The safety profile is consistent with what was previously reported for daratumumab or KRd, Dr Jakubowiak affirmed.

Serious TEAEs

Serious TEAEs occurred in 10 patients (46%), with many occurring in just 1 patient. Pulmonary embolism (PE) was the most frequent, occurring in 3 patients.

All patients were required to be on aspirin prophylaxis and 1 of the patients who had a PE discontinued therapy.

The number of patients with a serious TEAE reasonably related to an individual study drug were 3 (14%) for daratumumab, 5 (23%) for carfilzomib, 5 (23%) for lenalidomide, and 2 (9%) for dexamethasone.

The TEAEs of interest—tachycardia, congestive heart failure, and hypertension—occurred in a single patient each.

Overall, serious TEAEs were consistent with previous reports from KRd studies.

Echocardiogram assessment

Investigators conducted 30 systemic evaluations on the impact of this regimen on heart function.  The investigators observed no change from baseline through the duration of treatment in patients’ left ventricular ejection fractions.

One patient developed congestive heart failure, possibly related to daratumumab or carfilzomib. This patient resumed treatment with a reduced carfilzomib dose, elected ASCT on study day 113, and ended treatment with a VGPR.

“In all,” Dr Jakubowiak said, “we feel that there is no apparent signal of adverse impact of the addition of daratumumab on cardiac function.”

Infusion times and reactions

Overall, IRRs occurred in 27% of the patients, “which appears lower than with previous daratumumab studies,” Dr Jakubowiak noted. And IRRs occurred more frequently during the first infusion than subsequent infusions.

The split-dose infusion time was very similar to that of second and subsequent cycles.

There were limited events related to infusions. All were grade 1 or 2 and most occurred in only a single patient.

Response rate

The median number of treatment cycles administered was 11.5 (range, 2.0 – 13.0). The best response was 100% PR or better, 91% achieved VGPR or better, 42% CR or better, and 29% a stringent CR.

The depth of response improved with duration of treatment. For example, the sCR rate increased from 5% after 4 cycles to 29% at the end of treatment.

PFS was an exploratory endpoint. One patient progressed at 10.8 months and the 12-month PFS rate was 94% with all patients alive.

Stem cell harvest and ASCT

“For many of us,” Dr Jakubowiak commented, “it’s also of interest how this regimen will impact stem cell harvest.”

Nineteen of 22 patients were deemed to be transplant eligible, and the median number of CD34+ cells collected from them was 10.4 x 10cells/kg.

 

 

Patients had a median of 5 treatment cycles prior to stem cell harvest, and 14 (74%) had a VGPR or better prior to harvest.

The investigators believe stem cell yield was consistent with previous KRd studies.

Dr Jakubowiak commented that the deepening of response over time “is a phenomenon we think is important. . . . In all, the data from this small phase 1b study provide support for further evaluation of this regimen in newly diagnosed myeloma."

The study was funded by Janssen Research and Development, LLC. 

*Data presented during the meeting differ from the abstract.

Photo © ASCO/Scott Morgan 2017
Attendees at ASCO 2017 Annual Meeting

CHICAGO—Results of an open-label phase 1b study of daratumumab combined with carfilzomib, lenalidomide, and dexamethasone (KRd) in newly diagnosed multiple myeloma (MM) patients have shown the combination to be highly effective, with an overall response rate of 100%.

Ninety-one percent of patients achieved a very good partial response (VGPR) or better, and 43% achieved a complete response (CR) or better.

Investigators had hypothesized that rather than using autologous stem cell transplant (ASCT) to improve results of treatment with KRd, the combination could alternatively be improved by incorporating daratumumab into a KRd regimen.

Andrzej Jakubowiak, MD, of the University of Chicago Medical Center in Illinois, presented the findings of the MMY1001 study at the 2017 ASCO Annual Meeting (abstract 8000*).

“I think what was one of the more important developments in myeloma last year,” Dr Jakubowiak said, “was data from randomized studies showing that adding daratumumab to either lenalidomide and dexamethasone in the POLLUX study or bortezomib and dexamethasone, a proteasome inhibitor, in the CASTOR study, improves responses, depth of response, and . . .  dramatically improved progression-free survival.”

“[W]e have now the rationale to potentially combine daratumumab with both an IMiD and proteasome inhibitor,” he explained, “which led to the development of this phase 1b study in which we combined daratumumab with KRd and evaluated tolerability and efficacy.”

Study design

Twenty-two transplant-eligible or -ineligible newly diagnosed MM patients were enrolled on the study.

Treatment duration was planned to be 13 cycles or less and patients had the option to move to transplant after 4 cycles.

They could have no clinically significant cardiac disease and echocardiogram was required prior to transplant.

The dosing schedule was the established dosing schema for daratumumab and KRd with 2 notable differences in the 28-day cycles.

First, the daratumumab dose was a split dose. So patients received 8 mg/kg on days 1-2 of cycle 1, 16 mg/kg a week on cycle 2, 16 mg/kg every 2 weeks on cycles 3 – 6, and every 4th week thereafter.

The second difference was carfilzomib dosing was a weekly regimen with escalation from 20 mg/m2 on day 1, cycle 1 to 70 mg/m2 on day 8 of cycle 1.

Lenalidomide (25 mg on days 1-21 of each cycle) and dexamethasone (40 mg/week) were the standard regimens for these drugs.

The primary endpoint was safety and tolerability. The secondary endpoint was overall response rate (ORR), duration of response, time to response, and infusion-related reactions (IRR).

The study also had an exploratory endpoint of progression-free survival (PFS).

Baseline characteristics

Patients were a median age of 59.5 years (range 34 – 74). About two thirds were younger than 65 and one third were between 65 and 75.

A little over half were male and most (86%) were white.

A little more than half (55%) had an ECOG score of 0, 41% were ECOG 1, and 5% were ECOG 2.

Patient disposition

As of the cutoff date of March 24, 8 of the 22 patients enrolled (36%) discontinued treatment: 1 due to an adverse event (AE), 1 due to progressive disease, and 6 patients (27%) proceeded to ASCT.

Dr Jakubowiak pointed out that response was censored at this point for patients who proceeded to transplant.

The median follow-up was 10.8 months (range, 4.0 – 12.5) and the median number of treatment cycles was 11.5 (range, 1.0 – 13.0).

“What is of interest to many of us,” Dr Jakubowiak said, “is that patients were escalated to the planned dose of 70 mg/m2 by cycle 2 except for 3 patients.”

 

 

Of the 3, 1 discontinued before day 1 of the second cycle due to toxicity, 1 had a dose reduction to 56 mg/m2 at day of the second cycle, and 1 escalated to 70 mg/m2 at day 8 of cycle 3.

Ultimately, all patients who remained on study were able to escalate to 70 mg/m2.

Safety

The hematologic treatment-emergent adverse events (TEAE) generally followed what has been observed in similar studies before, Dr Jakubowiak noted.

Hematologic TEAEs of all grades occurring in 30% or more of patients were lymphopenia (68%), thrombocytopenia (55%), anemia (46%), leukopenia (41%), and neutropenia (32%).

The most common non-hematologic TEAEs of all grades occurring in 30% of patients or more were diarrhea (73%), upper respiratory infection (59%) cough, constipation, and fatigue (50% each), dyspnea and insomnia (46%), nausea, rash, and back pain (41%), muscle spasm (36%), and vomiting, pain in extremity, hyperglycemia, and increased ALT (32%).

The most common grade 3/4 TEAEs were infrequent and many events had none of grade 3/4 severity.

The safety profile is consistent with what was previously reported for daratumumab or KRd, Dr Jakubowiak affirmed.

Serious TEAEs

Serious TEAEs occurred in 10 patients (46%), with many occurring in just 1 patient. Pulmonary embolism (PE) was the most frequent, occurring in 3 patients.

All patients were required to be on aspirin prophylaxis and 1 of the patients who had a PE discontinued therapy.

The number of patients with a serious TEAE reasonably related to an individual study drug were 3 (14%) for daratumumab, 5 (23%) for carfilzomib, 5 (23%) for lenalidomide, and 2 (9%) for dexamethasone.

The TEAEs of interest—tachycardia, congestive heart failure, and hypertension—occurred in a single patient each.

Overall, serious TEAEs were consistent with previous reports from KRd studies.

Echocardiogram assessment

Investigators conducted 30 systemic evaluations on the impact of this regimen on heart function.  The investigators observed no change from baseline through the duration of treatment in patients’ left ventricular ejection fractions.

One patient developed congestive heart failure, possibly related to daratumumab or carfilzomib. This patient resumed treatment with a reduced carfilzomib dose, elected ASCT on study day 113, and ended treatment with a VGPR.

“In all,” Dr Jakubowiak said, “we feel that there is no apparent signal of adverse impact of the addition of daratumumab on cardiac function.”

Infusion times and reactions

Overall, IRRs occurred in 27% of the patients, “which appears lower than with previous daratumumab studies,” Dr Jakubowiak noted. And IRRs occurred more frequently during the first infusion than subsequent infusions.

The split-dose infusion time was very similar to that of second and subsequent cycles.

There were limited events related to infusions. All were grade 1 or 2 and most occurred in only a single patient.

Response rate

The median number of treatment cycles administered was 11.5 (range, 2.0 – 13.0). The best response was 100% PR or better, 91% achieved VGPR or better, 42% CR or better, and 29% a stringent CR.

The depth of response improved with duration of treatment. For example, the sCR rate increased from 5% after 4 cycles to 29% at the end of treatment.

PFS was an exploratory endpoint. One patient progressed at 10.8 months and the 12-month PFS rate was 94% with all patients alive.

Stem cell harvest and ASCT

“For many of us,” Dr Jakubowiak commented, “it’s also of interest how this regimen will impact stem cell harvest.”

Nineteen of 22 patients were deemed to be transplant eligible, and the median number of CD34+ cells collected from them was 10.4 x 10cells/kg.

 

 

Patients had a median of 5 treatment cycles prior to stem cell harvest, and 14 (74%) had a VGPR or better prior to harvest.

The investigators believe stem cell yield was consistent with previous KRd studies.

Dr Jakubowiak commented that the deepening of response over time “is a phenomenon we think is important. . . . In all, the data from this small phase 1b study provide support for further evaluation of this regimen in newly diagnosed myeloma."

The study was funded by Janssen Research and Development, LLC. 

*Data presented during the meeting differ from the abstract.

Publications
Publications
Topics
Article Type
Display Headline
Combo with daratumumab could be alternative to ASCT in MM
Display Headline
Combo with daratumumab could be alternative to ASCT in MM
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

ADT+RT duration can safely be shortened in high risk PC

Optimal treatment duration remains unclear
Article Type
Changed
Fri, 01/04/2019 - 13:37

 

– Androgen deprivation therapy (ADT) combined with radiation therapy can safely be reduced from 36 to 18 months without compromising outcomes or quality of life in patients with high-risk localized prostate cancer, according to the final results of a randomized phase III trial.

At a median of 9.4 year follow-up of 630 patients who were randomized to receive pelvic and prostate radiotherapy combined with either 36 or 18 months of ADT, the 10-year overall survival rate was 62.4% and 62.0% in the treatment arms, respectively (global hazard ratio, 1.024), Abdenour Nabid, MD, reported at the annual meeting of the American Society of Clinical Oncology.

Disease-free survival was 44.5% and 39.2% in the groups, respectively (HR, 0.835). This difference did not reach statistical significance, said Dr. Nabid of Centre Hospitalier Regional Universitaire, Sherbrooke, Quebec, Canada.

The disease-free survival curves separated over the course of the study, mainly because of a significant difference in biochemical failure between the groups, which favored the 36-month arm (24.8% vs. 31.0%; HR, 0.714), but this is not an unexpected finding with longer treatment, he explained.

“Does this biochemical control give you more control of the disease? I’m not sure,” he said, noting that bone metastases alone occurred in 23 and 24 patients in the 36 and 18 month treatment groups, respectively, and bone plus other site metastases occurred in 11 patients in each group. “At the end of the day, the P value (for disease-free survival) is not significant (.0768).”

Further, a quality of life analysis showed that patients in the 18-month treatment arm performed significantly better on 6 of 21 scales and 13 of 55 items addressing various quality of life factors. On two of these items – hot flushes and enjoyable sex – a clinically relevant difference of 10 or more points in mean scores was noted, he said.

Long-term ADT combined with radiotherapy is a standard treatment for patients with high-risk prostate cancer, but the optimal duration of treatment has not been defined, Dr. Nabid said.

The current trial looked at treatment duration in patients 80 years and younger (median of 71 in both groups) with T3-T4 disease, PSA levels greater than 20 mg/ml, and Gleason score greater than 7, with normal hepatic function and no regional disease or distant metastases. ADT included a 50 mg initial dose of bicalutamide daily for 1 month plus 10.8 mg of subcutaneous goserelin every 3 months, as well as pelvic and prostate radiotherapy.

On both univariate and multivariate analyses including age, Gleason score greater than 7, treatment duration, prostate-specific antigen greater than 20, T3-T4 disease, and biochemical failure during follow-up, only age and Gleason score were significantly associated with overall survival (HR, 1.05 for age in both analyses, and 1.40 and 1.42, respectively for Gleason score greater than 7 on univariate and multivariate analyses).

“In localized high-risk prostate cancer treated with radiotherapy and androgen deprivation therapy, androgen deprivation therapy duration can be safely reduced from 36 to 18 months,” Dr. Nabid said, adding that 18 months could represent a threshold effect in ADT duration and that side effects and treatment costs can be reduced with shorter duration of therapy.

“Eighteen months of ADT represents a new standard of care,” he concluded.

This study was funded by AstraZeneca Pharmaceuticals. Dr. Nabid has been a speaker, advisory board member, and/or received financial support from Janssen Canada, Sanofi, Astellas, and Bayer.

Body

 

Although it may seem reasonable to conclude based on the findings of this study that 18 months of ADT is similar to 36 months of ADT, the study was not designed to make this determination, according to Susan Halabi, PhD.

“In fact, a nonsignificant test result from a superiority comparison cannot be used to establish similarity,” Dr. Halabi said during a discussion of the findings at the meeting.

While she congratulated Dr. Nabid for his findings and long-term patient follow-up, she said questions remain.

“The optimal duration of ADT for high-risk localized prostate cancer is not known and remains a clinically important question,” she said.

Dr. Halabi is with Duke University, Durham, N.C. She reported consultant and/or advisory roles with Dendreon, Eisai, Genentech, Sanofi, and Tokai Pharmaceuticals and has received travel accommodations or expenses from Dendreon.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event
Related Articles
Body

 

Although it may seem reasonable to conclude based on the findings of this study that 18 months of ADT is similar to 36 months of ADT, the study was not designed to make this determination, according to Susan Halabi, PhD.

“In fact, a nonsignificant test result from a superiority comparison cannot be used to establish similarity,” Dr. Halabi said during a discussion of the findings at the meeting.

While she congratulated Dr. Nabid for his findings and long-term patient follow-up, she said questions remain.

“The optimal duration of ADT for high-risk localized prostate cancer is not known and remains a clinically important question,” she said.

Dr. Halabi is with Duke University, Durham, N.C. She reported consultant and/or advisory roles with Dendreon, Eisai, Genentech, Sanofi, and Tokai Pharmaceuticals and has received travel accommodations or expenses from Dendreon.

Body

 

Although it may seem reasonable to conclude based on the findings of this study that 18 months of ADT is similar to 36 months of ADT, the study was not designed to make this determination, according to Susan Halabi, PhD.

“In fact, a nonsignificant test result from a superiority comparison cannot be used to establish similarity,” Dr. Halabi said during a discussion of the findings at the meeting.

While she congratulated Dr. Nabid for his findings and long-term patient follow-up, she said questions remain.

“The optimal duration of ADT for high-risk localized prostate cancer is not known and remains a clinically important question,” she said.

Dr. Halabi is with Duke University, Durham, N.C. She reported consultant and/or advisory roles with Dendreon, Eisai, Genentech, Sanofi, and Tokai Pharmaceuticals and has received travel accommodations or expenses from Dendreon.

Title
Optimal treatment duration remains unclear
Optimal treatment duration remains unclear

 

– Androgen deprivation therapy (ADT) combined with radiation therapy can safely be reduced from 36 to 18 months without compromising outcomes or quality of life in patients with high-risk localized prostate cancer, according to the final results of a randomized phase III trial.

At a median of 9.4 year follow-up of 630 patients who were randomized to receive pelvic and prostate radiotherapy combined with either 36 or 18 months of ADT, the 10-year overall survival rate was 62.4% and 62.0% in the treatment arms, respectively (global hazard ratio, 1.024), Abdenour Nabid, MD, reported at the annual meeting of the American Society of Clinical Oncology.

Disease-free survival was 44.5% and 39.2% in the groups, respectively (HR, 0.835). This difference did not reach statistical significance, said Dr. Nabid of Centre Hospitalier Regional Universitaire, Sherbrooke, Quebec, Canada.

The disease-free survival curves separated over the course of the study, mainly because of a significant difference in biochemical failure between the groups, which favored the 36-month arm (24.8% vs. 31.0%; HR, 0.714), but this is not an unexpected finding with longer treatment, he explained.

“Does this biochemical control give you more control of the disease? I’m not sure,” he said, noting that bone metastases alone occurred in 23 and 24 patients in the 36 and 18 month treatment groups, respectively, and bone plus other site metastases occurred in 11 patients in each group. “At the end of the day, the P value (for disease-free survival) is not significant (.0768).”

Further, a quality of life analysis showed that patients in the 18-month treatment arm performed significantly better on 6 of 21 scales and 13 of 55 items addressing various quality of life factors. On two of these items – hot flushes and enjoyable sex – a clinically relevant difference of 10 or more points in mean scores was noted, he said.

Long-term ADT combined with radiotherapy is a standard treatment for patients with high-risk prostate cancer, but the optimal duration of treatment has not been defined, Dr. Nabid said.

The current trial looked at treatment duration in patients 80 years and younger (median of 71 in both groups) with T3-T4 disease, PSA levels greater than 20 mg/ml, and Gleason score greater than 7, with normal hepatic function and no regional disease or distant metastases. ADT included a 50 mg initial dose of bicalutamide daily for 1 month plus 10.8 mg of subcutaneous goserelin every 3 months, as well as pelvic and prostate radiotherapy.

On both univariate and multivariate analyses including age, Gleason score greater than 7, treatment duration, prostate-specific antigen greater than 20, T3-T4 disease, and biochemical failure during follow-up, only age and Gleason score were significantly associated with overall survival (HR, 1.05 for age in both analyses, and 1.40 and 1.42, respectively for Gleason score greater than 7 on univariate and multivariate analyses).

“In localized high-risk prostate cancer treated with radiotherapy and androgen deprivation therapy, androgen deprivation therapy duration can be safely reduced from 36 to 18 months,” Dr. Nabid said, adding that 18 months could represent a threshold effect in ADT duration and that side effects and treatment costs can be reduced with shorter duration of therapy.

“Eighteen months of ADT represents a new standard of care,” he concluded.

This study was funded by AstraZeneca Pharmaceuticals. Dr. Nabid has been a speaker, advisory board member, and/or received financial support from Janssen Canada, Sanofi, Astellas, and Bayer.

 

– Androgen deprivation therapy (ADT) combined with radiation therapy can safely be reduced from 36 to 18 months without compromising outcomes or quality of life in patients with high-risk localized prostate cancer, according to the final results of a randomized phase III trial.

At a median of 9.4 year follow-up of 630 patients who were randomized to receive pelvic and prostate radiotherapy combined with either 36 or 18 months of ADT, the 10-year overall survival rate was 62.4% and 62.0% in the treatment arms, respectively (global hazard ratio, 1.024), Abdenour Nabid, MD, reported at the annual meeting of the American Society of Clinical Oncology.

Disease-free survival was 44.5% and 39.2% in the groups, respectively (HR, 0.835). This difference did not reach statistical significance, said Dr. Nabid of Centre Hospitalier Regional Universitaire, Sherbrooke, Quebec, Canada.

The disease-free survival curves separated over the course of the study, mainly because of a significant difference in biochemical failure between the groups, which favored the 36-month arm (24.8% vs. 31.0%; HR, 0.714), but this is not an unexpected finding with longer treatment, he explained.

“Does this biochemical control give you more control of the disease? I’m not sure,” he said, noting that bone metastases alone occurred in 23 and 24 patients in the 36 and 18 month treatment groups, respectively, and bone plus other site metastases occurred in 11 patients in each group. “At the end of the day, the P value (for disease-free survival) is not significant (.0768).”

Further, a quality of life analysis showed that patients in the 18-month treatment arm performed significantly better on 6 of 21 scales and 13 of 55 items addressing various quality of life factors. On two of these items – hot flushes and enjoyable sex – a clinically relevant difference of 10 or more points in mean scores was noted, he said.

Long-term ADT combined with radiotherapy is a standard treatment for patients with high-risk prostate cancer, but the optimal duration of treatment has not been defined, Dr. Nabid said.

The current trial looked at treatment duration in patients 80 years and younger (median of 71 in both groups) with T3-T4 disease, PSA levels greater than 20 mg/ml, and Gleason score greater than 7, with normal hepatic function and no regional disease or distant metastases. ADT included a 50 mg initial dose of bicalutamide daily for 1 month plus 10.8 mg of subcutaneous goserelin every 3 months, as well as pelvic and prostate radiotherapy.

On both univariate and multivariate analyses including age, Gleason score greater than 7, treatment duration, prostate-specific antigen greater than 20, T3-T4 disease, and biochemical failure during follow-up, only age and Gleason score were significantly associated with overall survival (HR, 1.05 for age in both analyses, and 1.40 and 1.42, respectively for Gleason score greater than 7 on univariate and multivariate analyses).

“In localized high-risk prostate cancer treated with radiotherapy and androgen deprivation therapy, androgen deprivation therapy duration can be safely reduced from 36 to 18 months,” Dr. Nabid said, adding that 18 months could represent a threshold effect in ADT duration and that side effects and treatment costs can be reduced with shorter duration of therapy.

“Eighteen months of ADT represents a new standard of care,” he concluded.

This study was funded by AstraZeneca Pharmaceuticals. Dr. Nabid has been a speaker, advisory board member, and/or received financial support from Janssen Canada, Sanofi, Astellas, and Bayer.

Publications
Publications
Topics
Article Type
Sections
Article Source

AT THE 2017 ASCO ANNUAL MEETING

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

 

Key clinical point: ADT combined with radiation therapy can safely be reduced from 36 to 18 months in patients with high-risk localized prostate cancer.

Major finding: Ten-year overall survival was 62.4% and 62.0% for patients randomized to receive pelvic and prostate radiotherapy combined with either 36 or 18 months of androgen deprivation therapy, respectively (global hazard ratio, 1.024).

Data source: A randomized phase III trial of 630 patients.

Disclosures: This study was funded by AstraZeneca Pharmaceuticals. Dr. Nabid has been a speaker, advisory board member, and/or received financial support from Janssen Canada, Sanofi, Astellas, and Bayer.

Novel CAR T cells drive high objective response rate in multiple myeloma

Article Type
Changed
Fri, 01/04/2019 - 10:04

 

CHICAGO – CARs just keep getting better: In an early clinical trial, a chimeric antigen receptor (CAR) T-cell construct targeting B-cell maturation protein induced clinical remissions in 33 of 35 patients with relapsed/refractory multiple myeloma who were treated in an early clinical trial.

Wanhong Zhao, MD, from the Second Affiliated Hospital of Xi’an Jiaotong University in China
Dr. Wanhong Zhao
He presented early results from a single-arm trial of the CAR T cell, labeled LCAR-B38M, in 35 patients at a briefing at the American Society of Clinical Oncology (ASCO) annual meeting.

“I think what you’re seeing here is the expansion of immunotherapy to cancers that really are refractory to chemotherapy and how immunotherapy is now providing hope to a lot of patients with cancers that were not really responding to our standard chemotherapies,” commented ASCO expert Michael S. Sabel, MD, of the University of Michigan, Ann Arbor. “What I also think is really fascinating about this and similar forms of research is that you are now seeing the merger of immunotherapy with personalized medicine.”

Current CAR T-cell technologies targeting CD19 or a similar antigen have shown efficacy against acute lymphoblastic leukemia and some forms of lymphoma, but it has been difficult to identify a suitable target in multiple myeloma.

B-cell maturation antigen (BCMA) was first described in myeloma in 2004 as a mechanism for the growth and survival of malignant plasma cells.

Several research groups are currently investigating CAR T cells or monoclonal antibodies targeted to BCMA.

In the study by Dr. Zhao and his colleagues, 19 patients had been followed for more than 4 months before the data cutoff in January 2017. Four months is the minimum established by the International Myeloma Working Group for efficacy assessment.

Of the 19 patients, 14 had achieved a stringent complete response (sCR), 4 had very good partial responses, and 1 had a partial response, for an objective response rate of 100%.

No patients who achieved an sCR have had relapses, and all five patients who have been in follow-up for more than a year have maintained their sCRs and are free of minimal residual disease, Dr. Zhao reported.

One patient with a very good partial response had disease progression, with recurrence of an extramedullary lesion that had previously disappeared.

The most common adverse event was cytokine release syndrome, which occurred in 85% of patients, but the condition was transient and manageable in a majority, Dr. Zhao said.

Two patients developed grade 3 cytokine release syndrome and were treated with tocilizumab (Actemra).

The investigators plan to enroll a total of 100 patients from participating hospitals in China and are planning a U.S. trial for launch in early 2018.

The investigators hope to look at BCMA CAR-T cell therapy in the frontline for patients with newly diagnosed multiple myeloma.

The study was funded by Legend Biotech. Coauthor Fran (Xiaohu) Fan, MD, PhD, is employed by the company. Dr. Zhao did not report disclosures. Dr. Sabel had no disclosures relevant to the study.

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

 

CHICAGO – CARs just keep getting better: In an early clinical trial, a chimeric antigen receptor (CAR) T-cell construct targeting B-cell maturation protein induced clinical remissions in 33 of 35 patients with relapsed/refractory multiple myeloma who were treated in an early clinical trial.

Wanhong Zhao, MD, from the Second Affiliated Hospital of Xi’an Jiaotong University in China
Dr. Wanhong Zhao
He presented early results from a single-arm trial of the CAR T cell, labeled LCAR-B38M, in 35 patients at a briefing at the American Society of Clinical Oncology (ASCO) annual meeting.

“I think what you’re seeing here is the expansion of immunotherapy to cancers that really are refractory to chemotherapy and how immunotherapy is now providing hope to a lot of patients with cancers that were not really responding to our standard chemotherapies,” commented ASCO expert Michael S. Sabel, MD, of the University of Michigan, Ann Arbor. “What I also think is really fascinating about this and similar forms of research is that you are now seeing the merger of immunotherapy with personalized medicine.”

Current CAR T-cell technologies targeting CD19 or a similar antigen have shown efficacy against acute lymphoblastic leukemia and some forms of lymphoma, but it has been difficult to identify a suitable target in multiple myeloma.

B-cell maturation antigen (BCMA) was first described in myeloma in 2004 as a mechanism for the growth and survival of malignant plasma cells.

Several research groups are currently investigating CAR T cells or monoclonal antibodies targeted to BCMA.

In the study by Dr. Zhao and his colleagues, 19 patients had been followed for more than 4 months before the data cutoff in January 2017. Four months is the minimum established by the International Myeloma Working Group for efficacy assessment.

Of the 19 patients, 14 had achieved a stringent complete response (sCR), 4 had very good partial responses, and 1 had a partial response, for an objective response rate of 100%.

No patients who achieved an sCR have had relapses, and all five patients who have been in follow-up for more than a year have maintained their sCRs and are free of minimal residual disease, Dr. Zhao reported.

One patient with a very good partial response had disease progression, with recurrence of an extramedullary lesion that had previously disappeared.

The most common adverse event was cytokine release syndrome, which occurred in 85% of patients, but the condition was transient and manageable in a majority, Dr. Zhao said.

Two patients developed grade 3 cytokine release syndrome and were treated with tocilizumab (Actemra).

The investigators plan to enroll a total of 100 patients from participating hospitals in China and are planning a U.S. trial for launch in early 2018.

The investigators hope to look at BCMA CAR-T cell therapy in the frontline for patients with newly diagnosed multiple myeloma.

The study was funded by Legend Biotech. Coauthor Fran (Xiaohu) Fan, MD, PhD, is employed by the company. Dr. Zhao did not report disclosures. Dr. Sabel had no disclosures relevant to the study.

 

CHICAGO – CARs just keep getting better: In an early clinical trial, a chimeric antigen receptor (CAR) T-cell construct targeting B-cell maturation protein induced clinical remissions in 33 of 35 patients with relapsed/refractory multiple myeloma who were treated in an early clinical trial.

Wanhong Zhao, MD, from the Second Affiliated Hospital of Xi’an Jiaotong University in China
Dr. Wanhong Zhao
He presented early results from a single-arm trial of the CAR T cell, labeled LCAR-B38M, in 35 patients at a briefing at the American Society of Clinical Oncology (ASCO) annual meeting.

“I think what you’re seeing here is the expansion of immunotherapy to cancers that really are refractory to chemotherapy and how immunotherapy is now providing hope to a lot of patients with cancers that were not really responding to our standard chemotherapies,” commented ASCO expert Michael S. Sabel, MD, of the University of Michigan, Ann Arbor. “What I also think is really fascinating about this and similar forms of research is that you are now seeing the merger of immunotherapy with personalized medicine.”

Current CAR T-cell technologies targeting CD19 or a similar antigen have shown efficacy against acute lymphoblastic leukemia and some forms of lymphoma, but it has been difficult to identify a suitable target in multiple myeloma.

B-cell maturation antigen (BCMA) was first described in myeloma in 2004 as a mechanism for the growth and survival of malignant plasma cells.

Several research groups are currently investigating CAR T cells or monoclonal antibodies targeted to BCMA.

In the study by Dr. Zhao and his colleagues, 19 patients had been followed for more than 4 months before the data cutoff in January 2017. Four months is the minimum established by the International Myeloma Working Group for efficacy assessment.

Of the 19 patients, 14 had achieved a stringent complete response (sCR), 4 had very good partial responses, and 1 had a partial response, for an objective response rate of 100%.

No patients who achieved an sCR have had relapses, and all five patients who have been in follow-up for more than a year have maintained their sCRs and are free of minimal residual disease, Dr. Zhao reported.

One patient with a very good partial response had disease progression, with recurrence of an extramedullary lesion that had previously disappeared.

The most common adverse event was cytokine release syndrome, which occurred in 85% of patients, but the condition was transient and manageable in a majority, Dr. Zhao said.

Two patients developed grade 3 cytokine release syndrome and were treated with tocilizumab (Actemra).

The investigators plan to enroll a total of 100 patients from participating hospitals in China and are planning a U.S. trial for launch in early 2018.

The investigators hope to look at BCMA CAR-T cell therapy in the frontline for patients with newly diagnosed multiple myeloma.

The study was funded by Legend Biotech. Coauthor Fran (Xiaohu) Fan, MD, PhD, is employed by the company. Dr. Zhao did not report disclosures. Dr. Sabel had no disclosures relevant to the study.

Publications
Publications
Topics
Article Type
Sections
Article Source

AT THE 2017 ASCO ANNUAL MEETING

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

 

Key clinical point: All of 19 patients treated with the CAR T-cell construct targeting B-cell maturation antigen had an objective response.

Major finding: Of 35 patients with relapsed/refractory multiple myeloma treated with BCMA, 33 had remissions.

Data source: A prospective single-arm study of 35 patients, with enrollment planned for 100.

Disclosures: The study was funded by Legend Biotech. Coauthor Fran (Xiaohu) Fan, MD, PhD, is employed by the company. Dr. Zhao did not report disclosures. Dr. Sabel had no disclosures relevant to the study.

T-DMI doesn’t wow in HER2-overexpressing NSCLC

Article Type
Changed
Fri, 01/04/2019 - 13:37

 

–Beyond its role in advanced breast cancer, the antibody-drug conjugate ado-trastuzumab-emtansine (T-DM1; Kadcyla) showed modest activity in some patients with metastatic non–small cell lung cancer (mNSCLC) overexpressing the human epidermal growth factor-2 (HER2) receptor, investigators reported.

In an ongoing phase II study, 4 of 20 patients with mNSCLC whose tumors expressed the highest levels of HER2 had partial responses. In contrast, none of the 20 patients with tumors overexpressing HER2 at lower levels had responses, reported Thomas E. Stinchcombe, MD, of Duke University, Durham, N.C.

Dr. Thomas E Stinchcombe of Duke University, Durham, N.C.
Dr. Thomas E. Stinchcombe
“Additional investigation into improved detection of HER2 amplification and other biomarkers may help to refine the patient population that is likely to benefit from T-DM1,” he said at the annual meeting of the American Society of Clinical Oncology.

Previous studies have shown that HER2 overexpression by immunohistochemistry (IHC) is associated with poor prognosis in patients with NSCLC, but in contrast to breast and gastric cancers, HER2 overexpression in NSCLC is not always accompanied by HER2 amplifications.

“HER2 amplifications and HER2 mutations are generally mutually exclusive in NSCLC. Given the known mechanism of action of T-DM1, HER2 overexpression was chosen as an inclusion criterion for this study,” Dr. Stinchcombe said.

They enrolled patients with HER2-overexpressing mNSCLC who had disease progression following platinum-based chemotherapy. The patients were assigned to one of two 20-patient cohorts based on IHC2+ (10% or more of cells stained with 2+ intensity), or IHC3+ (10% or more of cells stained with 3+intensity).

For the primary endpoint of treatment response none of the patients in the IHC2+ cohort had objective responses by Response Evaluation Criteria in Solid Tumors (RECIST), although eight patients in this cohort had stable disease, including one patient who remained in stable disease status on treatment out to 21 months at last follow-up.

In the IHC3+ cohort, four patients had partial responses, for an objective response rate of 20%. The median duration of response was 7.3 months. Two patients in this cohort had stable disease.

Median PFS was similar between the cohorts, at 2.6 months for IHC2+ and 2.7 months for IHC3+. The respective median OS durations were 12.2 and 12.1 months.

The safety profile of T-DM1 in this population was similar to that seen in breast cancer. There were two grade 3 serious adverse events: one infusion-related hypersensitivity reaction, and one case of thrombocytopenia. There were 10 grade 3 events of any kind, 1 grade 4 event, and 1 treatment withdrawal due to a grade 2 infusion reaction.

Dr. Leena Gandhi of New York University Langone School of Medicine in Manhattan
Dr. Leena Gandhi
The unremarkable findings from this study raise the question of whether HER2 is the right target in this population, said Leena Gandhi, MD, PhD, of New York University Langone School of Medicine in Manhattan, the invited discussant.

“I think it’s honestly a little hard to tell from this study, which is very small, whether there is really a role of [HER2] overexpression in actually driving oncogenesis and being a target for T-DM1,” she said.

Dr. Stinchcombe reported institutional research funding from Hoffmann-LaRoche, which sponsored the trial, and several coauthors are employees of the company.

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

 

–Beyond its role in advanced breast cancer, the antibody-drug conjugate ado-trastuzumab-emtansine (T-DM1; Kadcyla) showed modest activity in some patients with metastatic non–small cell lung cancer (mNSCLC) overexpressing the human epidermal growth factor-2 (HER2) receptor, investigators reported.

In an ongoing phase II study, 4 of 20 patients with mNSCLC whose tumors expressed the highest levels of HER2 had partial responses. In contrast, none of the 20 patients with tumors overexpressing HER2 at lower levels had responses, reported Thomas E. Stinchcombe, MD, of Duke University, Durham, N.C.

Dr. Thomas E Stinchcombe of Duke University, Durham, N.C.
Dr. Thomas E. Stinchcombe
“Additional investigation into improved detection of HER2 amplification and other biomarkers may help to refine the patient population that is likely to benefit from T-DM1,” he said at the annual meeting of the American Society of Clinical Oncology.

Previous studies have shown that HER2 overexpression by immunohistochemistry (IHC) is associated with poor prognosis in patients with NSCLC, but in contrast to breast and gastric cancers, HER2 overexpression in NSCLC is not always accompanied by HER2 amplifications.

“HER2 amplifications and HER2 mutations are generally mutually exclusive in NSCLC. Given the known mechanism of action of T-DM1, HER2 overexpression was chosen as an inclusion criterion for this study,” Dr. Stinchcombe said.

They enrolled patients with HER2-overexpressing mNSCLC who had disease progression following platinum-based chemotherapy. The patients were assigned to one of two 20-patient cohorts based on IHC2+ (10% or more of cells stained with 2+ intensity), or IHC3+ (10% or more of cells stained with 3+intensity).

For the primary endpoint of treatment response none of the patients in the IHC2+ cohort had objective responses by Response Evaluation Criteria in Solid Tumors (RECIST), although eight patients in this cohort had stable disease, including one patient who remained in stable disease status on treatment out to 21 months at last follow-up.

In the IHC3+ cohort, four patients had partial responses, for an objective response rate of 20%. The median duration of response was 7.3 months. Two patients in this cohort had stable disease.

Median PFS was similar between the cohorts, at 2.6 months for IHC2+ and 2.7 months for IHC3+. The respective median OS durations were 12.2 and 12.1 months.

The safety profile of T-DM1 in this population was similar to that seen in breast cancer. There were two grade 3 serious adverse events: one infusion-related hypersensitivity reaction, and one case of thrombocytopenia. There were 10 grade 3 events of any kind, 1 grade 4 event, and 1 treatment withdrawal due to a grade 2 infusion reaction.

Dr. Leena Gandhi of New York University Langone School of Medicine in Manhattan
Dr. Leena Gandhi
The unremarkable findings from this study raise the question of whether HER2 is the right target in this population, said Leena Gandhi, MD, PhD, of New York University Langone School of Medicine in Manhattan, the invited discussant.

“I think it’s honestly a little hard to tell from this study, which is very small, whether there is really a role of [HER2] overexpression in actually driving oncogenesis and being a target for T-DM1,” she said.

Dr. Stinchcombe reported institutional research funding from Hoffmann-LaRoche, which sponsored the trial, and several coauthors are employees of the company.

 

–Beyond its role in advanced breast cancer, the antibody-drug conjugate ado-trastuzumab-emtansine (T-DM1; Kadcyla) showed modest activity in some patients with metastatic non–small cell lung cancer (mNSCLC) overexpressing the human epidermal growth factor-2 (HER2) receptor, investigators reported.

In an ongoing phase II study, 4 of 20 patients with mNSCLC whose tumors expressed the highest levels of HER2 had partial responses. In contrast, none of the 20 patients with tumors overexpressing HER2 at lower levels had responses, reported Thomas E. Stinchcombe, MD, of Duke University, Durham, N.C.

Dr. Thomas E Stinchcombe of Duke University, Durham, N.C.
Dr. Thomas E. Stinchcombe
“Additional investigation into improved detection of HER2 amplification and other biomarkers may help to refine the patient population that is likely to benefit from T-DM1,” he said at the annual meeting of the American Society of Clinical Oncology.

Previous studies have shown that HER2 overexpression by immunohistochemistry (IHC) is associated with poor prognosis in patients with NSCLC, but in contrast to breast and gastric cancers, HER2 overexpression in NSCLC is not always accompanied by HER2 amplifications.

“HER2 amplifications and HER2 mutations are generally mutually exclusive in NSCLC. Given the known mechanism of action of T-DM1, HER2 overexpression was chosen as an inclusion criterion for this study,” Dr. Stinchcombe said.

They enrolled patients with HER2-overexpressing mNSCLC who had disease progression following platinum-based chemotherapy. The patients were assigned to one of two 20-patient cohorts based on IHC2+ (10% or more of cells stained with 2+ intensity), or IHC3+ (10% or more of cells stained with 3+intensity).

For the primary endpoint of treatment response none of the patients in the IHC2+ cohort had objective responses by Response Evaluation Criteria in Solid Tumors (RECIST), although eight patients in this cohort had stable disease, including one patient who remained in stable disease status on treatment out to 21 months at last follow-up.

In the IHC3+ cohort, four patients had partial responses, for an objective response rate of 20%. The median duration of response was 7.3 months. Two patients in this cohort had stable disease.

Median PFS was similar between the cohorts, at 2.6 months for IHC2+ and 2.7 months for IHC3+. The respective median OS durations were 12.2 and 12.1 months.

The safety profile of T-DM1 in this population was similar to that seen in breast cancer. There were two grade 3 serious adverse events: one infusion-related hypersensitivity reaction, and one case of thrombocytopenia. There were 10 grade 3 events of any kind, 1 grade 4 event, and 1 treatment withdrawal due to a grade 2 infusion reaction.

Dr. Leena Gandhi of New York University Langone School of Medicine in Manhattan
Dr. Leena Gandhi
The unremarkable findings from this study raise the question of whether HER2 is the right target in this population, said Leena Gandhi, MD, PhD, of New York University Langone School of Medicine in Manhattan, the invited discussant.

“I think it’s honestly a little hard to tell from this study, which is very small, whether there is really a role of [HER2] overexpression in actually driving oncogenesis and being a target for T-DM1,” she said.

Dr. Stinchcombe reported institutional research funding from Hoffmann-LaRoche, which sponsored the trial, and several coauthors are employees of the company.

Publications
Publications
Topics
Article Type
Sections
Article Source

AT ASCO 2017

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

 

Key clinical point: Some non–small cell lung cancer (NSCLC) tumors overexpress HER2, suggesting the use of T-DM1, an anti-HER2 antibody drug conjugate.

Major finding: Four of 20 patients with HER2-overexpressing NSCLC has partial responses to T-DM1 therapy.

Data source: Open-label prospective trial of 40 patients with NSCLC positive for HER2.

Disclosures: Dr. Stinchcombe reported institutional research funding from Hoffmann-LaRoche, which sponsored the trial, and several coauthors are employees of the company.

VIDEO: Immune therapy effective, durable in treatment-naive melanoma brain metastases

Article Type
Changed
Mon, 01/14/2019 - 10:03

 

CHICAGO– Immune therapy shows promise for use in the treatment of melanoma brain metastases, especially for treatment-naive patients, judging from the findings of a new phase II randomized study.

For patients with asymptomatic brain metastases from melanoma who had not had prior treatment, nivolumab combined with ipilimumab produced a 50% intracranial response rate after at least 12 weeks of therapy. When nivolumab alone was given to untreated patients, the intracranial response rate was 21%, Georgina Long MD, PhD, co–medical director of the Melanoma Institute Australia , said during a video interview at the annual meeting of the American Society of Clinical Oncology.

“If you look at progression-free survival by response, none of our complete responders have progressed,” said Dr. Long. “And this is with a median follow-up of 16.4 months.” The partial responders have also done well, with little progression, she said. “Remember, these patients usually survive only a few weeks.”

The Anti-PD1 Brain Collaboration study, a phase II clinical trial, enrolled patients with melanoma brain metastases at least 5 mm but less than 40 mm in diameter who had not received previous anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4), anti-programmed cell death protein 1 (anti-PD-1), or anti-programmed death-ligand 1 (anti-PD-L1) therapies. Patients were permitted to have had previous BRAF and MEK inhibitor therapies. Asymptomatic patients who had no previous local brain therapy (i.e., radiation treatment or surgery) were randomized 1:1 to receive nivolumab alone, or nivolumab plus ipilimumab.

The nivolumab arm received 3 mg/kg by intravenous infusion every 2 weeks. The combination arm began with nivolumab 1 mg/kg and ipilimumab 3 mg/kg every 3 weeks for four doses. After this, they also received nivolumab 3 mg/kg monotherapy every 2 weeks.

The third cohort – a small group of 15 patients who received nivolumab alone – either had symptomatic brain metastases or leptomeningeal disease and could have had previous brain surgery or radiotherapy. Unlike the first two cohorts, they were also permitted to be on up to 10 mg/day of prednisone; these patients received nivolumab alone at 3 mg/kg every 2 weeks.

For all patients, immune therapy was given until the disease progressed, consent was withdrawn, or patients experienced unacceptable toxicity or they died.

“We were most interested in the randomized cohorts,” said Dr. Long. Interestingly, she said, ipilimumab became available in Australia when 27 patients were enrolled in the nivolumab arm and 26 to the combination arm. “So we stopped the monotherapy arm, and the rest of the 60 patients to be recruited all went into the combination arm,” she said. A total of 76 patients were recruited, 33 into the combination arm, 27 to the asymptomatic nivolumab monotherapy arm, and 16 to the symptomatic and/or previously treated arm.

Data analysis from the point of the data cut included 67 patients who were followed for a period ranging from 5 to 34 months. Intracranial disease was evaluated by gadolinium-enhanced MRI and modified Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria.

“The results of the trial were very interesting,” said Dr. Long. The nivolumab plus ipilimumab combo resulted in an overall 42% intracranial response rate, while nivolumab alone produced an overall intracranial response rate of 21%. However, patients in either arm who had prior BRAF or MEK inhibitor exposure “didn’t do too well on immunotherapy,” said Dr. Long, noting that the response rate was just 16% for these patients. These were, she said, “small numbers, but still, an interesting signal there.”

When comparing the secondary endpoint of extracranial response to intracranial response on a per-patient basis, Dr. Long and her collaborators could see that “the intracranial and extracranial results were mostly concordant.”

Analysis of the additional secondary endpoints of progression-free survival (PFS) and overall survival (OS) also showed an interesting pattern, said Dr. Long. After an initial drop-off period of about 5 months, the curves for patients in all arms have stabilized, so that patients who were responders are maintaining that response. The overall 6-month PFS rate for the combination cohort was 47%, with a durable response: “If you look at the curve, it’s flattened out since that stage, and we haven’t had any progression since that time,” said Dr. Long. The PFS rate was 29% for the cohort receiving nivolumab alone. “Activity is highest when nivolumab and ipilimumab are given upfront,” said Dr. Long.

For asymptomatic patients pretreated with BRAF or MEK inhibitors, “activity is low,” said Dr. Long, with an intracranial response rate of 16% in both cohorts.

Symptomatic patients who were more heavily pretreated fared even worse: “The activity of nivolumab monotherapy is low after multiple modality therapy or in leptomeningeal melanoma,” said Dr. Long. The intracranial response rate in the third cohort was just 6%.

The combination therapy cohort had the most treatment-related adverse events, with 96% of patients experiencing some adverse event. About half (12/26, 46%) had grade 3 or 4 events, and the same number had a serious adverse event. Seven patients (27%) discontinued therapy because of treatment-related adverse events in the combination study arm. However, said Dr. Long, this side effect profile is in keeping with what has been seen in other studies of combination therapy with nivolumab and ipilimumab. “There were not unexpected adverse events,” she said.

Dr. Long reported relationships with Bristol-Myers Squibb, Merck, and Roche.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
 

 

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

 

CHICAGO– Immune therapy shows promise for use in the treatment of melanoma brain metastases, especially for treatment-naive patients, judging from the findings of a new phase II randomized study.

For patients with asymptomatic brain metastases from melanoma who had not had prior treatment, nivolumab combined with ipilimumab produced a 50% intracranial response rate after at least 12 weeks of therapy. When nivolumab alone was given to untreated patients, the intracranial response rate was 21%, Georgina Long MD, PhD, co–medical director of the Melanoma Institute Australia , said during a video interview at the annual meeting of the American Society of Clinical Oncology.

“If you look at progression-free survival by response, none of our complete responders have progressed,” said Dr. Long. “And this is with a median follow-up of 16.4 months.” The partial responders have also done well, with little progression, she said. “Remember, these patients usually survive only a few weeks.”

The Anti-PD1 Brain Collaboration study, a phase II clinical trial, enrolled patients with melanoma brain metastases at least 5 mm but less than 40 mm in diameter who had not received previous anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4), anti-programmed cell death protein 1 (anti-PD-1), or anti-programmed death-ligand 1 (anti-PD-L1) therapies. Patients were permitted to have had previous BRAF and MEK inhibitor therapies. Asymptomatic patients who had no previous local brain therapy (i.e., radiation treatment or surgery) were randomized 1:1 to receive nivolumab alone, or nivolumab plus ipilimumab.

The nivolumab arm received 3 mg/kg by intravenous infusion every 2 weeks. The combination arm began with nivolumab 1 mg/kg and ipilimumab 3 mg/kg every 3 weeks for four doses. After this, they also received nivolumab 3 mg/kg monotherapy every 2 weeks.

The third cohort – a small group of 15 patients who received nivolumab alone – either had symptomatic brain metastases or leptomeningeal disease and could have had previous brain surgery or radiotherapy. Unlike the first two cohorts, they were also permitted to be on up to 10 mg/day of prednisone; these patients received nivolumab alone at 3 mg/kg every 2 weeks.

For all patients, immune therapy was given until the disease progressed, consent was withdrawn, or patients experienced unacceptable toxicity or they died.

“We were most interested in the randomized cohorts,” said Dr. Long. Interestingly, she said, ipilimumab became available in Australia when 27 patients were enrolled in the nivolumab arm and 26 to the combination arm. “So we stopped the monotherapy arm, and the rest of the 60 patients to be recruited all went into the combination arm,” she said. A total of 76 patients were recruited, 33 into the combination arm, 27 to the asymptomatic nivolumab monotherapy arm, and 16 to the symptomatic and/or previously treated arm.

Data analysis from the point of the data cut included 67 patients who were followed for a period ranging from 5 to 34 months. Intracranial disease was evaluated by gadolinium-enhanced MRI and modified Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria.

“The results of the trial were very interesting,” said Dr. Long. The nivolumab plus ipilimumab combo resulted in an overall 42% intracranial response rate, while nivolumab alone produced an overall intracranial response rate of 21%. However, patients in either arm who had prior BRAF or MEK inhibitor exposure “didn’t do too well on immunotherapy,” said Dr. Long, noting that the response rate was just 16% for these patients. These were, she said, “small numbers, but still, an interesting signal there.”

When comparing the secondary endpoint of extracranial response to intracranial response on a per-patient basis, Dr. Long and her collaborators could see that “the intracranial and extracranial results were mostly concordant.”

Analysis of the additional secondary endpoints of progression-free survival (PFS) and overall survival (OS) also showed an interesting pattern, said Dr. Long. After an initial drop-off period of about 5 months, the curves for patients in all arms have stabilized, so that patients who were responders are maintaining that response. The overall 6-month PFS rate for the combination cohort was 47%, with a durable response: “If you look at the curve, it’s flattened out since that stage, and we haven’t had any progression since that time,” said Dr. Long. The PFS rate was 29% for the cohort receiving nivolumab alone. “Activity is highest when nivolumab and ipilimumab are given upfront,” said Dr. Long.

For asymptomatic patients pretreated with BRAF or MEK inhibitors, “activity is low,” said Dr. Long, with an intracranial response rate of 16% in both cohorts.

Symptomatic patients who were more heavily pretreated fared even worse: “The activity of nivolumab monotherapy is low after multiple modality therapy or in leptomeningeal melanoma,” said Dr. Long. The intracranial response rate in the third cohort was just 6%.

The combination therapy cohort had the most treatment-related adverse events, with 96% of patients experiencing some adverse event. About half (12/26, 46%) had grade 3 or 4 events, and the same number had a serious adverse event. Seven patients (27%) discontinued therapy because of treatment-related adverse events in the combination study arm. However, said Dr. Long, this side effect profile is in keeping with what has been seen in other studies of combination therapy with nivolumab and ipilimumab. “There were not unexpected adverse events,” she said.

Dr. Long reported relationships with Bristol-Myers Squibb, Merck, and Roche.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
 

 

 

CHICAGO– Immune therapy shows promise for use in the treatment of melanoma brain metastases, especially for treatment-naive patients, judging from the findings of a new phase II randomized study.

For patients with asymptomatic brain metastases from melanoma who had not had prior treatment, nivolumab combined with ipilimumab produced a 50% intracranial response rate after at least 12 weeks of therapy. When nivolumab alone was given to untreated patients, the intracranial response rate was 21%, Georgina Long MD, PhD, co–medical director of the Melanoma Institute Australia , said during a video interview at the annual meeting of the American Society of Clinical Oncology.

“If you look at progression-free survival by response, none of our complete responders have progressed,” said Dr. Long. “And this is with a median follow-up of 16.4 months.” The partial responders have also done well, with little progression, she said. “Remember, these patients usually survive only a few weeks.”

The Anti-PD1 Brain Collaboration study, a phase II clinical trial, enrolled patients with melanoma brain metastases at least 5 mm but less than 40 mm in diameter who had not received previous anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4), anti-programmed cell death protein 1 (anti-PD-1), or anti-programmed death-ligand 1 (anti-PD-L1) therapies. Patients were permitted to have had previous BRAF and MEK inhibitor therapies. Asymptomatic patients who had no previous local brain therapy (i.e., radiation treatment or surgery) were randomized 1:1 to receive nivolumab alone, or nivolumab plus ipilimumab.

The nivolumab arm received 3 mg/kg by intravenous infusion every 2 weeks. The combination arm began with nivolumab 1 mg/kg and ipilimumab 3 mg/kg every 3 weeks for four doses. After this, they also received nivolumab 3 mg/kg monotherapy every 2 weeks.

The third cohort – a small group of 15 patients who received nivolumab alone – either had symptomatic brain metastases or leptomeningeal disease and could have had previous brain surgery or radiotherapy. Unlike the first two cohorts, they were also permitted to be on up to 10 mg/day of prednisone; these patients received nivolumab alone at 3 mg/kg every 2 weeks.

For all patients, immune therapy was given until the disease progressed, consent was withdrawn, or patients experienced unacceptable toxicity or they died.

“We were most interested in the randomized cohorts,” said Dr. Long. Interestingly, she said, ipilimumab became available in Australia when 27 patients were enrolled in the nivolumab arm and 26 to the combination arm. “So we stopped the monotherapy arm, and the rest of the 60 patients to be recruited all went into the combination arm,” she said. A total of 76 patients were recruited, 33 into the combination arm, 27 to the asymptomatic nivolumab monotherapy arm, and 16 to the symptomatic and/or previously treated arm.

Data analysis from the point of the data cut included 67 patients who were followed for a period ranging from 5 to 34 months. Intracranial disease was evaluated by gadolinium-enhanced MRI and modified Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria.

“The results of the trial were very interesting,” said Dr. Long. The nivolumab plus ipilimumab combo resulted in an overall 42% intracranial response rate, while nivolumab alone produced an overall intracranial response rate of 21%. However, patients in either arm who had prior BRAF or MEK inhibitor exposure “didn’t do too well on immunotherapy,” said Dr. Long, noting that the response rate was just 16% for these patients. These were, she said, “small numbers, but still, an interesting signal there.”

When comparing the secondary endpoint of extracranial response to intracranial response on a per-patient basis, Dr. Long and her collaborators could see that “the intracranial and extracranial results were mostly concordant.”

Analysis of the additional secondary endpoints of progression-free survival (PFS) and overall survival (OS) also showed an interesting pattern, said Dr. Long. After an initial drop-off period of about 5 months, the curves for patients in all arms have stabilized, so that patients who were responders are maintaining that response. The overall 6-month PFS rate for the combination cohort was 47%, with a durable response: “If you look at the curve, it’s flattened out since that stage, and we haven’t had any progression since that time,” said Dr. Long. The PFS rate was 29% for the cohort receiving nivolumab alone. “Activity is highest when nivolumab and ipilimumab are given upfront,” said Dr. Long.

For asymptomatic patients pretreated with BRAF or MEK inhibitors, “activity is low,” said Dr. Long, with an intracranial response rate of 16% in both cohorts.

Symptomatic patients who were more heavily pretreated fared even worse: “The activity of nivolumab monotherapy is low after multiple modality therapy or in leptomeningeal melanoma,” said Dr. Long. The intracranial response rate in the third cohort was just 6%.

The combination therapy cohort had the most treatment-related adverse events, with 96% of patients experiencing some adverse event. About half (12/26, 46%) had grade 3 or 4 events, and the same number had a serious adverse event. Seven patients (27%) discontinued therapy because of treatment-related adverse events in the combination study arm. However, said Dr. Long, this side effect profile is in keeping with what has been seen in other studies of combination therapy with nivolumab and ipilimumab. “There were not unexpected adverse events,” she said.

Dr. Long reported relationships with Bristol-Myers Squibb, Merck, and Roche.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
 

 

Publications
Publications
Topics
Article Type
Sections
Article Source

AT ASCO 2017

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

Deep molecular responses achievable in AML pts treated with gilteritinib

Article Type
Changed
Mon, 06/12/2017 - 01:00
Display Headline
Deep molecular responses achievable in AML pts treated with gilteritinib

Photo © ASCO/Danny Morton 2017
Lobby of McCormick Place during ASCO 2017

CHICAGO—Next generation sequencing (NGS) has shown that the FLT3 inhibitor gilteritinib can produce deep molecular responses in a subset of patients with acute myeloid leukemia (AML), according to new research.

Gilteritinib is a highly selective FLT3/AXL inhibitor that is active against FLT3-ITD and FLT3-D835 mutations, but minimal residual disease (MRD) had not systematically been assessed previously in AML patients treated with potent FLT3 inhibitors.

Investigators believed that MRD evaluation in these patients could serve as a useful marker of FLT3 inhibitor efficacy. They therefore conducted an exploratory analysis of a subset of AML patients treated with gilteritinib on the Chrysalis study.

Jessica K. Altman, MD, of the Robert H. Lurie Cancer Center of Northwestern University in Chicago, Illinois, presented the findings at the ASCO 2017 Annual Meeting (abstract 7003).

Chrysalis study: Efficacy and survival

The phase 1/2 Chrysalis study examined the tolerability and antileukemic activity of once daily gilteritinib in a FLT3-ITD-enriched relapsed/refractory AML population of approximately 250 patients.

Overall, gilteritinib was well tolerated and had consistency and potent FLT3 inhibition at doses of >80 mg/day.

The maximum tolerated dose was 300 mg/day. Dose-limiting toxicities were diarrhea and liver function abnormalities.

The greatest overall response rate was 52% and the longest median overall survival (OS) duration was 31 weeks, observed in patients at doses >80 mg/day.

The composite complete remission (CR) rate, comprised of CR, CR with incomplete count recovery (Cri), and CR with incomplete platelet recovery (CRp), was 41%.

The median OS was 31 weeks, and median duration of response 20 weeks.

“Survival probabilities demonstrated that the overall survival for patients who received 80 mg of gilteritinib was higher than those who received less than 80 mg,” Dr Altman said.

Molecular response assessment

Dr Altman then presented the molecular response assessment.

The investigators included all FLT3-ITD mutated patients enrolled in the gilteritinib 120 and 200 mg/day dose cohorts and had bone marrow aspirates available at baseline and at 1 or more additional time points.

“The group I’m reporting on,” Dr Altman explained, “comprises 51% of all FLT3-ITD mutated patients treated at these 2 dose levels.”

FLT3-ITD and total FLT3 alleles were quantified by a novel NGS assay using an Illumina® sequencing platform. Read depth of at least 100,000 reads per sample were implemented.

“Evaluation of MRD was exploratory and it was not prespecified in the study,” Dr Altman noted.

Hence, the investigators defined a molecular response as an ITD signal ratio—FLT3-ITD : FLT3 total—of <10-2.

They defined major molecular response (MMR) as an ITD signal ratio of <10-3, and negative MRD status as <10-4.

Patient characteristics

Baseline characteristics of the 80 patients in the MRD analysis group were similar to those of the entire Chrysalis study population.

Median age was 61 years (range, 23 – 86) and the patients were heavily pretreated: 35% had 3 or more prior lines of AML therapy, and 28% had received a prior FLT3 inhibitor. About a third had prior allogeneic hematopoietic stem cell transplant.

Molecular response

Median OS in this cohort was 32.6 weeks, very similar to the entire study population.

Twenty patients (25%) achieved a molecular response, 18 (23%) an MMR, and 13 (16%) were MRD negative.

The median time to achieve a minimum ITD signal ratio was 8.2 weeks (range, 3.7 – 64).

And molecular response correlated with improved OS.

“The 20 patients who achieved a molecular response had a median overall survival of 59.6 weeks,” Dr Altman said, “which is statistically significantly different and I think clinically different than those who did not attain a molecular response.”

 

 

Patients who did not achieve a molecular response had a median overall survival of 28.4 weeks.

“As you could predict,” she added, “the molecular response was greater in those who attained a complete remission than those who had a CRp or Cri.”

Investigators observed similar results in patients who achieved an MMR, using the the cutoff point of 10-3.

“When we stratified by MRD negative status,” she said, “which was an ITD signal ratio of 10-4 or better, there’s clear separation of the Kaplan Meier curves for OS in this cohort again.”

Dr Altman pointed out that this was the first clinical trial to demonstrate that patients with AML treated with a FLT3 inhibitor can attain a molecular response.

“Also, and importantly, there is now a sensitive and specific assay for the detection of minimal residual disease in FLT3-ITD mutated patients and it has the potential to be widely adopted across trials and in clinical practice.”

MRD is prospectively being evaluated in 2 gilteritinib phase 3 maintenance studies.

The trial was sponsored by Astellas Pharma Global Development, Inc. 

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

Photo © ASCO/Danny Morton 2017
Lobby of McCormick Place during ASCO 2017

CHICAGO—Next generation sequencing (NGS) has shown that the FLT3 inhibitor gilteritinib can produce deep molecular responses in a subset of patients with acute myeloid leukemia (AML), according to new research.

Gilteritinib is a highly selective FLT3/AXL inhibitor that is active against FLT3-ITD and FLT3-D835 mutations, but minimal residual disease (MRD) had not systematically been assessed previously in AML patients treated with potent FLT3 inhibitors.

Investigators believed that MRD evaluation in these patients could serve as a useful marker of FLT3 inhibitor efficacy. They therefore conducted an exploratory analysis of a subset of AML patients treated with gilteritinib on the Chrysalis study.

Jessica K. Altman, MD, of the Robert H. Lurie Cancer Center of Northwestern University in Chicago, Illinois, presented the findings at the ASCO 2017 Annual Meeting (abstract 7003).

Chrysalis study: Efficacy and survival

The phase 1/2 Chrysalis study examined the tolerability and antileukemic activity of once daily gilteritinib in a FLT3-ITD-enriched relapsed/refractory AML population of approximately 250 patients.

Overall, gilteritinib was well tolerated and had consistency and potent FLT3 inhibition at doses of >80 mg/day.

The maximum tolerated dose was 300 mg/day. Dose-limiting toxicities were diarrhea and liver function abnormalities.

The greatest overall response rate was 52% and the longest median overall survival (OS) duration was 31 weeks, observed in patients at doses >80 mg/day.

The composite complete remission (CR) rate, comprised of CR, CR with incomplete count recovery (Cri), and CR with incomplete platelet recovery (CRp), was 41%.

The median OS was 31 weeks, and median duration of response 20 weeks.

“Survival probabilities demonstrated that the overall survival for patients who received 80 mg of gilteritinib was higher than those who received less than 80 mg,” Dr Altman said.

Molecular response assessment

Dr Altman then presented the molecular response assessment.

The investigators included all FLT3-ITD mutated patients enrolled in the gilteritinib 120 and 200 mg/day dose cohorts and had bone marrow aspirates available at baseline and at 1 or more additional time points.

“The group I’m reporting on,” Dr Altman explained, “comprises 51% of all FLT3-ITD mutated patients treated at these 2 dose levels.”

FLT3-ITD and total FLT3 alleles were quantified by a novel NGS assay using an Illumina® sequencing platform. Read depth of at least 100,000 reads per sample were implemented.

“Evaluation of MRD was exploratory and it was not prespecified in the study,” Dr Altman noted.

Hence, the investigators defined a molecular response as an ITD signal ratio—FLT3-ITD : FLT3 total—of <10-2.

They defined major molecular response (MMR) as an ITD signal ratio of <10-3, and negative MRD status as <10-4.

Patient characteristics

Baseline characteristics of the 80 patients in the MRD analysis group were similar to those of the entire Chrysalis study population.

Median age was 61 years (range, 23 – 86) and the patients were heavily pretreated: 35% had 3 or more prior lines of AML therapy, and 28% had received a prior FLT3 inhibitor. About a third had prior allogeneic hematopoietic stem cell transplant.

Molecular response

Median OS in this cohort was 32.6 weeks, very similar to the entire study population.

Twenty patients (25%) achieved a molecular response, 18 (23%) an MMR, and 13 (16%) were MRD negative.

The median time to achieve a minimum ITD signal ratio was 8.2 weeks (range, 3.7 – 64).

And molecular response correlated with improved OS.

“The 20 patients who achieved a molecular response had a median overall survival of 59.6 weeks,” Dr Altman said, “which is statistically significantly different and I think clinically different than those who did not attain a molecular response.”

 

 

Patients who did not achieve a molecular response had a median overall survival of 28.4 weeks.

“As you could predict,” she added, “the molecular response was greater in those who attained a complete remission than those who had a CRp or Cri.”

Investigators observed similar results in patients who achieved an MMR, using the the cutoff point of 10-3.

“When we stratified by MRD negative status,” she said, “which was an ITD signal ratio of 10-4 or better, there’s clear separation of the Kaplan Meier curves for OS in this cohort again.”

Dr Altman pointed out that this was the first clinical trial to demonstrate that patients with AML treated with a FLT3 inhibitor can attain a molecular response.

“Also, and importantly, there is now a sensitive and specific assay for the detection of minimal residual disease in FLT3-ITD mutated patients and it has the potential to be widely adopted across trials and in clinical practice.”

MRD is prospectively being evaluated in 2 gilteritinib phase 3 maintenance studies.

The trial was sponsored by Astellas Pharma Global Development, Inc. 

Photo © ASCO/Danny Morton 2017
Lobby of McCormick Place during ASCO 2017

CHICAGO—Next generation sequencing (NGS) has shown that the FLT3 inhibitor gilteritinib can produce deep molecular responses in a subset of patients with acute myeloid leukemia (AML), according to new research.

Gilteritinib is a highly selective FLT3/AXL inhibitor that is active against FLT3-ITD and FLT3-D835 mutations, but minimal residual disease (MRD) had not systematically been assessed previously in AML patients treated with potent FLT3 inhibitors.

Investigators believed that MRD evaluation in these patients could serve as a useful marker of FLT3 inhibitor efficacy. They therefore conducted an exploratory analysis of a subset of AML patients treated with gilteritinib on the Chrysalis study.

Jessica K. Altman, MD, of the Robert H. Lurie Cancer Center of Northwestern University in Chicago, Illinois, presented the findings at the ASCO 2017 Annual Meeting (abstract 7003).

Chrysalis study: Efficacy and survival

The phase 1/2 Chrysalis study examined the tolerability and antileukemic activity of once daily gilteritinib in a FLT3-ITD-enriched relapsed/refractory AML population of approximately 250 patients.

Overall, gilteritinib was well tolerated and had consistency and potent FLT3 inhibition at doses of >80 mg/day.

The maximum tolerated dose was 300 mg/day. Dose-limiting toxicities were diarrhea and liver function abnormalities.

The greatest overall response rate was 52% and the longest median overall survival (OS) duration was 31 weeks, observed in patients at doses >80 mg/day.

The composite complete remission (CR) rate, comprised of CR, CR with incomplete count recovery (Cri), and CR with incomplete platelet recovery (CRp), was 41%.

The median OS was 31 weeks, and median duration of response 20 weeks.

“Survival probabilities demonstrated that the overall survival for patients who received 80 mg of gilteritinib was higher than those who received less than 80 mg,” Dr Altman said.

Molecular response assessment

Dr Altman then presented the molecular response assessment.

The investigators included all FLT3-ITD mutated patients enrolled in the gilteritinib 120 and 200 mg/day dose cohorts and had bone marrow aspirates available at baseline and at 1 or more additional time points.

“The group I’m reporting on,” Dr Altman explained, “comprises 51% of all FLT3-ITD mutated patients treated at these 2 dose levels.”

FLT3-ITD and total FLT3 alleles were quantified by a novel NGS assay using an Illumina® sequencing platform. Read depth of at least 100,000 reads per sample were implemented.

“Evaluation of MRD was exploratory and it was not prespecified in the study,” Dr Altman noted.

Hence, the investigators defined a molecular response as an ITD signal ratio—FLT3-ITD : FLT3 total—of <10-2.

They defined major molecular response (MMR) as an ITD signal ratio of <10-3, and negative MRD status as <10-4.

Patient characteristics

Baseline characteristics of the 80 patients in the MRD analysis group were similar to those of the entire Chrysalis study population.

Median age was 61 years (range, 23 – 86) and the patients were heavily pretreated: 35% had 3 or more prior lines of AML therapy, and 28% had received a prior FLT3 inhibitor. About a third had prior allogeneic hematopoietic stem cell transplant.

Molecular response

Median OS in this cohort was 32.6 weeks, very similar to the entire study population.

Twenty patients (25%) achieved a molecular response, 18 (23%) an MMR, and 13 (16%) were MRD negative.

The median time to achieve a minimum ITD signal ratio was 8.2 weeks (range, 3.7 – 64).

And molecular response correlated with improved OS.

“The 20 patients who achieved a molecular response had a median overall survival of 59.6 weeks,” Dr Altman said, “which is statistically significantly different and I think clinically different than those who did not attain a molecular response.”

 

 

Patients who did not achieve a molecular response had a median overall survival of 28.4 weeks.

“As you could predict,” she added, “the molecular response was greater in those who attained a complete remission than those who had a CRp or Cri.”

Investigators observed similar results in patients who achieved an MMR, using the the cutoff point of 10-3.

“When we stratified by MRD negative status,” she said, “which was an ITD signal ratio of 10-4 or better, there’s clear separation of the Kaplan Meier curves for OS in this cohort again.”

Dr Altman pointed out that this was the first clinical trial to demonstrate that patients with AML treated with a FLT3 inhibitor can attain a molecular response.

“Also, and importantly, there is now a sensitive and specific assay for the detection of minimal residual disease in FLT3-ITD mutated patients and it has the potential to be widely adopted across trials and in clinical practice.”

MRD is prospectively being evaluated in 2 gilteritinib phase 3 maintenance studies.

The trial was sponsored by Astellas Pharma Global Development, Inc. 

Publications
Publications
Topics
Article Type
Display Headline
Deep molecular responses achievable in AML pts treated with gilteritinib
Display Headline
Deep molecular responses achievable in AML pts treated with gilteritinib
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Majority of AML patients do not receive recommended molecular genetic testing

Article Type
Changed
Sun, 06/11/2017 - 01:00
Display Headline
Majority of AML patients do not receive recommended molecular genetic testing

DNA helices Image courtesy of the National Institute of General Medical Sciences
General Medical Sciences
DNA helices Image courtesy of the National Institute of

CHICAGO—While 67% of newly diagnosed patients with acute myeloid leukemia (AML) receive some genetic testing, only 9% receive all 7 of the genetic tests recommended by the National Comprehensive Cancer Network (NCCN), according to new research.

The data comes from the CONNECT MDS/AML Disease Registry, which collects treatment and outcome statistics from 86 sites across the United States, both academic medical centers and community settings. The findings reflect data gathered from 2013 to 2016.

Previously, data on genetic testing in AML patients came primarily from clinical trials, where adherence to guidelines is very high. The current analysis evaluated adherence to genetic testing guidelines in AML treated outside the clinic trial setting.

Daniel A. Pollyea, MD, of the University of Colorado Comprehensive Cancer Center in Aurora, presented the findings on behalf of the CONNECT MDS/AML Disease Registry Scientific Steering Committee (abstract 7022).

The CONNECT registry is a US prospective, observational cohort study of patients with newly diagnosed AML or myelodysplastic syndromes (MDS) aged 55 years or older. Enrollment is ongoing and study clinicians make all clinical decisions.

NCCN guidelines recommend testing AML patients for NPM1, FLT3-ITD, CEBPA, IDH1, IDH2, DNMT3A, and KIT mutations.

“We now know a tremendous amount about the genetic underpinnings of the disease,” Dr Pollyea said.

“We can test for these genetic changes in the clinic to see what’s making a patient’s disease tick. And often there are targeted therapies that can be matched with these genetic changes," he added. "But there’s a disconnect between what can be done, what should be done, and what is being done.”

The current analysis evaluated genetic testing in 259 AML patients, 173 (67%) of whom had some genetic testing.

The likelihood of patients getting tested varied by type of treatment center, age, karyotype, and insurance.

Patients treated at academic medical centers had higher rates of testing than those treated at community clinics (76% and 62%, respectively), P= 0.018

Patients younger than 65 years more often were tested than older patients (83% and 60%, respectively), P= 0.0003.

Patients with non-Medicare insurance were more often tested than those with Medicare (74% and 61%, respectively), P= .025.

And patients with normal karyotype were more often tested than those with abnormal karyotype (77% and 59%, respectively), P= 0.006.

Of the 173 patients who had some genetic testing, only 15 (9%) had all the molecular tests recommended by NCCN.

Of the 7 recommended tests, NPM1 (77%) and FLT3-ITD (76%) were most often reported and DNMT3A least often (16%).

Dr Pollyea attributed the lack of adherence to the guidelines in part to willingness of insurance companies to pay for testing.

He also suggested the guidelines themselves might need adjustment, given the low adherence rate.

Nevertheless, he affirmed the guidelines are well founded. “I think the guidelines are pretty solid and, in my opinion, I would say they don’t go far enough in recommending genetic testing.”

“We’re in our infancy with this testing, and even earlier than infancy in seeing how we’re doing on testing. But now with this registry we at least have the infrastructure available to ask these kinds of questions,” Dr Pollyea added. 

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

DNA helices Image courtesy of the National Institute of General Medical Sciences
General Medical Sciences
DNA helices Image courtesy of the National Institute of

CHICAGO—While 67% of newly diagnosed patients with acute myeloid leukemia (AML) receive some genetic testing, only 9% receive all 7 of the genetic tests recommended by the National Comprehensive Cancer Network (NCCN), according to new research.

The data comes from the CONNECT MDS/AML Disease Registry, which collects treatment and outcome statistics from 86 sites across the United States, both academic medical centers and community settings. The findings reflect data gathered from 2013 to 2016.

Previously, data on genetic testing in AML patients came primarily from clinical trials, where adherence to guidelines is very high. The current analysis evaluated adherence to genetic testing guidelines in AML treated outside the clinic trial setting.

Daniel A. Pollyea, MD, of the University of Colorado Comprehensive Cancer Center in Aurora, presented the findings on behalf of the CONNECT MDS/AML Disease Registry Scientific Steering Committee (abstract 7022).

The CONNECT registry is a US prospective, observational cohort study of patients with newly diagnosed AML or myelodysplastic syndromes (MDS) aged 55 years or older. Enrollment is ongoing and study clinicians make all clinical decisions.

NCCN guidelines recommend testing AML patients for NPM1, FLT3-ITD, CEBPA, IDH1, IDH2, DNMT3A, and KIT mutations.

“We now know a tremendous amount about the genetic underpinnings of the disease,” Dr Pollyea said.

“We can test for these genetic changes in the clinic to see what’s making a patient’s disease tick. And often there are targeted therapies that can be matched with these genetic changes," he added. "But there’s a disconnect between what can be done, what should be done, and what is being done.”

The current analysis evaluated genetic testing in 259 AML patients, 173 (67%) of whom had some genetic testing.

The likelihood of patients getting tested varied by type of treatment center, age, karyotype, and insurance.

Patients treated at academic medical centers had higher rates of testing than those treated at community clinics (76% and 62%, respectively), P= 0.018

Patients younger than 65 years more often were tested than older patients (83% and 60%, respectively), P= 0.0003.

Patients with non-Medicare insurance were more often tested than those with Medicare (74% and 61%, respectively), P= .025.

And patients with normal karyotype were more often tested than those with abnormal karyotype (77% and 59%, respectively), P= 0.006.

Of the 173 patients who had some genetic testing, only 15 (9%) had all the molecular tests recommended by NCCN.

Of the 7 recommended tests, NPM1 (77%) and FLT3-ITD (76%) were most often reported and DNMT3A least often (16%).

Dr Pollyea attributed the lack of adherence to the guidelines in part to willingness of insurance companies to pay for testing.

He also suggested the guidelines themselves might need adjustment, given the low adherence rate.

Nevertheless, he affirmed the guidelines are well founded. “I think the guidelines are pretty solid and, in my opinion, I would say they don’t go far enough in recommending genetic testing.”

“We’re in our infancy with this testing, and even earlier than infancy in seeing how we’re doing on testing. But now with this registry we at least have the infrastructure available to ask these kinds of questions,” Dr Pollyea added. 

DNA helices Image courtesy of the National Institute of General Medical Sciences
General Medical Sciences
DNA helices Image courtesy of the National Institute of

CHICAGO—While 67% of newly diagnosed patients with acute myeloid leukemia (AML) receive some genetic testing, only 9% receive all 7 of the genetic tests recommended by the National Comprehensive Cancer Network (NCCN), according to new research.

The data comes from the CONNECT MDS/AML Disease Registry, which collects treatment and outcome statistics from 86 sites across the United States, both academic medical centers and community settings. The findings reflect data gathered from 2013 to 2016.

Previously, data on genetic testing in AML patients came primarily from clinical trials, where adherence to guidelines is very high. The current analysis evaluated adherence to genetic testing guidelines in AML treated outside the clinic trial setting.

Daniel A. Pollyea, MD, of the University of Colorado Comprehensive Cancer Center in Aurora, presented the findings on behalf of the CONNECT MDS/AML Disease Registry Scientific Steering Committee (abstract 7022).

The CONNECT registry is a US prospective, observational cohort study of patients with newly diagnosed AML or myelodysplastic syndromes (MDS) aged 55 years or older. Enrollment is ongoing and study clinicians make all clinical decisions.

NCCN guidelines recommend testing AML patients for NPM1, FLT3-ITD, CEBPA, IDH1, IDH2, DNMT3A, and KIT mutations.

“We now know a tremendous amount about the genetic underpinnings of the disease,” Dr Pollyea said.

“We can test for these genetic changes in the clinic to see what’s making a patient’s disease tick. And often there are targeted therapies that can be matched with these genetic changes," he added. "But there’s a disconnect between what can be done, what should be done, and what is being done.”

The current analysis evaluated genetic testing in 259 AML patients, 173 (67%) of whom had some genetic testing.

The likelihood of patients getting tested varied by type of treatment center, age, karyotype, and insurance.

Patients treated at academic medical centers had higher rates of testing than those treated at community clinics (76% and 62%, respectively), P= 0.018

Patients younger than 65 years more often were tested than older patients (83% and 60%, respectively), P= 0.0003.

Patients with non-Medicare insurance were more often tested than those with Medicare (74% and 61%, respectively), P= .025.

And patients with normal karyotype were more often tested than those with abnormal karyotype (77% and 59%, respectively), P= 0.006.

Of the 173 patients who had some genetic testing, only 15 (9%) had all the molecular tests recommended by NCCN.

Of the 7 recommended tests, NPM1 (77%) and FLT3-ITD (76%) were most often reported and DNMT3A least often (16%).

Dr Pollyea attributed the lack of adherence to the guidelines in part to willingness of insurance companies to pay for testing.

He also suggested the guidelines themselves might need adjustment, given the low adherence rate.

Nevertheless, he affirmed the guidelines are well founded. “I think the guidelines are pretty solid and, in my opinion, I would say they don’t go far enough in recommending genetic testing.”

“We’re in our infancy with this testing, and even earlier than infancy in seeing how we’re doing on testing. But now with this registry we at least have the infrastructure available to ask these kinds of questions,” Dr Pollyea added. 

Publications
Publications
Topics
Article Type
Display Headline
Majority of AML patients do not receive recommended molecular genetic testing
Display Headline
Majority of AML patients do not receive recommended molecular genetic testing
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Dasatinib potentially a new SOC for children with CML-CP

Article Type
Changed
Fri, 06/09/2017 - 01:00
Display Headline
Dasatinib potentially a new SOC for children with CML-CP

Child with cancer Photo by Bill Branson
Photo by Bill Branson
Child with cancer

CHICAGO—The largest ongoing and prospective trial of pediatric patients with chronic myeloid leukemia in chronic phase (CML-CP), according to the best knowledge of the investigators, has found dasatinib to be safe and effective as first- or second-line therapy for these children.

Patients refractory to or intolerant of imatinib had a major cytogenetic response (MCyR) by 3 months and responses at 12 and 24 months exceeded 90%.

Newly diagnosed patients had a complete cytogenetic response (CCyR) by 6 months.

“We believe our data suggests that dasatinib could be considered as a new standard of care (SOC) for children with CML in chronic phase,” said study author Lia Gore, MD, of the University of Colorado School of Medicine/Children’s Hospital Colorado in Aurora.

She presented the findings of the study at the ASCO 2017 Annual Meeting (abstract 10511).

Study design

CA 180-226 is a phase 2, open-label, nonrandomized, prospective study conducted in 18 countries. Patients younger than 18 years with newly diagnosed CML-CP, or imatinib-resistant/intolerant (R/I) CML-CP, or CML in accelerated phase, or Ph+ acute lymphoblastic leukemia (ALL) were enrolled on the study between March 2009 and September 2014.

Dr Gore’s presentation focused on the CML-CP patients in the study, both the newly diagnosed and the imatinib-R/I patients.

The imatinib-R/I patients received dasatinib 60 mg/m2 tablets once daily, and the newly diagnosed patients received the same tablet dosage daily or a powdered formulation for oral suspension (PFOS) of dasatinib at 72 mg/m2 daily.

Dr Gore noted the different dosage in the oral suspension formulation is based on bioavailability studies performed in adults, which was determined to be equivalent to the 60 mg/m2 tablet formulation.

Once accrual was reached in the tablet cohort, newly diagnosed patients were accrued to the PFOS cohort. The patients on PFOS could switch to tablets after a year or more on the oral suspension.

Patients remained on treatment until disease progression, unacceptable toxicity occurred, or the patient/physician preference.

All patients had a minimum follow-up of 2 years. The longest follow-up was more than 90 months.

Primary objectives of the study were MCyR greater than 30% for imatinib-R/I patients and complete CCyR greater than 55% for newly diagnosed patients.

Secondary objectives included time to and duration of response, major molecular response (MMR), progression-free survival (PFS), overall survival (OS), and safety.

Baseline patient characteristics

One hundred thirteen patients were treated across the 3 cohorts—29 in the imatinib-R/I receiving tablets, 51 newly diagnosed patients receiving tablets, and 33 newly diagnosed patients in the PFOS arm. A total of 84 patients had newly diagnosed disease.

Of the 29 imatinib-R/I patients, 25 were resistant, 2 intolerant, and 2 undetermined. And 6 of the 25 resistant patients had defined imatinib-resistance mutations.

Median age was 13.8 years in the imatinib-R/I cohort, 12.9 years in the newly diagnosed on tablets, and 11.7 years in the PFOS group. Other baseline characteristics were similar among the cohorts.

“Importantly, there were 3 patients less than 2 years of age, and a substantial proportion of patients were actually less than 12 years of age,” Dr Gore pointed out.

Dasatinib exposure

The median duration of therapy was 50 months and 42 months in the R/I and newly diagnosed cohorts, respectively.

Forty-eight percent of the imatinib-R/I patients and 73% of the newly diagnosed patients are still on treatment. A relatively small number of patients discontinued therapy.

The median duration of therapy was shorter in the PFOS cohort because they were enrolled only after accrual to the tablet cohort. However, they were also followed up for more than 2 years.

 

 

Of the 33 patients on PFOS, 22 eventually switched to tablet formulation.

Results

The primary endpoint for imatinib-R/I patients—MCyR greater than 30%—was reached by 3 months, and MCyR at 12 and 24 months exceeded 90%. The median time to response was 3.1 months (range, 2.8 – 4.1), and median duration of response was not yet reached (range, 54.9 – not estimable).

For newly diagnosed patients, the preset defined rate of interest of 55% for CCyR was reached as early as 6 months, and exceeded 90% by 12 and 24 months.

Dr Gore pointed out that intolerant patients also reached CCyR relatively quickly, although it was not a specified endpoint.

Data indicate that responses occurred relatively quickly and continued to increase over time of follow-up.

MMR also continued to increase over time and showed no difference between formulation and response rate.

Median PFS has not been reached, as only 7 patients in each cohort had disease progression.

One imatinib-R/I patient died 1 year after stopping treatment. The patient, who had a GI bleed unrelated to dasatinib, had discontinued therapy for progressive disease with loss of MCyR.

Safety

Overall safety was very similar to the dasatinib exposure and experience in adults, Dr Gore said, and there were no differences in events between PFOS and tablets.

One patient in the PFOS cohort had a dasatinib-related grade 3 hypersensitivity reaction, which resolved after discontinuation of dasatinib.

“What’s important here,” she said, “is that there were almost no adverse events of severity in either cohort, only 1 in the imatinib refractory and intolerant and 1 in the newly diagnosed cohorts.”

“Most importantly for those of us with a lot of experience in this field,” she added, “there were no occurrences of pleural effusion, pericardial effusion, pulmonary edema, pulmonary hypertension, or any vascular occlusive events in patients noted on this trial.”

“Additionally, for pediatricians, we care a lot about what happens to growth in these patients and prospectively we collected a lot of data related to growth parameters in bone growth and development."

Of the dasatinib-related adverse events occurring in 10% or more of patients, there were only 5 growth and development events noted out of the 113 patients treated and all were grade 1 or 2 events, Dr Gore pointed out.

In the R/I cohort, one patient had osteopenia and gynecomastia. At the time of data analysis, this event had resolved even though the patient continued on dasatinib.

“We believe our data suggests that dasatinib could be considered as a new standard of care for children with CML in chronic phase,” she said.

“It includes the advantage of a liquid formulation as well as the advantages of once daily dosing and administration without regard to fed or fasting state,” she added, “which for all of us who treat children know could be quite important.”

The study was funded by Bristol-Myers Squibb.

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

Child with cancer Photo by Bill Branson
Photo by Bill Branson
Child with cancer

CHICAGO—The largest ongoing and prospective trial of pediatric patients with chronic myeloid leukemia in chronic phase (CML-CP), according to the best knowledge of the investigators, has found dasatinib to be safe and effective as first- or second-line therapy for these children.

Patients refractory to or intolerant of imatinib had a major cytogenetic response (MCyR) by 3 months and responses at 12 and 24 months exceeded 90%.

Newly diagnosed patients had a complete cytogenetic response (CCyR) by 6 months.

“We believe our data suggests that dasatinib could be considered as a new standard of care (SOC) for children with CML in chronic phase,” said study author Lia Gore, MD, of the University of Colorado School of Medicine/Children’s Hospital Colorado in Aurora.

She presented the findings of the study at the ASCO 2017 Annual Meeting (abstract 10511).

Study design

CA 180-226 is a phase 2, open-label, nonrandomized, prospective study conducted in 18 countries. Patients younger than 18 years with newly diagnosed CML-CP, or imatinib-resistant/intolerant (R/I) CML-CP, or CML in accelerated phase, or Ph+ acute lymphoblastic leukemia (ALL) were enrolled on the study between March 2009 and September 2014.

Dr Gore’s presentation focused on the CML-CP patients in the study, both the newly diagnosed and the imatinib-R/I patients.

The imatinib-R/I patients received dasatinib 60 mg/m2 tablets once daily, and the newly diagnosed patients received the same tablet dosage daily or a powdered formulation for oral suspension (PFOS) of dasatinib at 72 mg/m2 daily.

Dr Gore noted the different dosage in the oral suspension formulation is based on bioavailability studies performed in adults, which was determined to be equivalent to the 60 mg/m2 tablet formulation.

Once accrual was reached in the tablet cohort, newly diagnosed patients were accrued to the PFOS cohort. The patients on PFOS could switch to tablets after a year or more on the oral suspension.

Patients remained on treatment until disease progression, unacceptable toxicity occurred, or the patient/physician preference.

All patients had a minimum follow-up of 2 years. The longest follow-up was more than 90 months.

Primary objectives of the study were MCyR greater than 30% for imatinib-R/I patients and complete CCyR greater than 55% for newly diagnosed patients.

Secondary objectives included time to and duration of response, major molecular response (MMR), progression-free survival (PFS), overall survival (OS), and safety.

Baseline patient characteristics

One hundred thirteen patients were treated across the 3 cohorts—29 in the imatinib-R/I receiving tablets, 51 newly diagnosed patients receiving tablets, and 33 newly diagnosed patients in the PFOS arm. A total of 84 patients had newly diagnosed disease.

Of the 29 imatinib-R/I patients, 25 were resistant, 2 intolerant, and 2 undetermined. And 6 of the 25 resistant patients had defined imatinib-resistance mutations.

Median age was 13.8 years in the imatinib-R/I cohort, 12.9 years in the newly diagnosed on tablets, and 11.7 years in the PFOS group. Other baseline characteristics were similar among the cohorts.

“Importantly, there were 3 patients less than 2 years of age, and a substantial proportion of patients were actually less than 12 years of age,” Dr Gore pointed out.

Dasatinib exposure

The median duration of therapy was 50 months and 42 months in the R/I and newly diagnosed cohorts, respectively.

Forty-eight percent of the imatinib-R/I patients and 73% of the newly diagnosed patients are still on treatment. A relatively small number of patients discontinued therapy.

The median duration of therapy was shorter in the PFOS cohort because they were enrolled only after accrual to the tablet cohort. However, they were also followed up for more than 2 years.

 

 

Of the 33 patients on PFOS, 22 eventually switched to tablet formulation.

Results

The primary endpoint for imatinib-R/I patients—MCyR greater than 30%—was reached by 3 months, and MCyR at 12 and 24 months exceeded 90%. The median time to response was 3.1 months (range, 2.8 – 4.1), and median duration of response was not yet reached (range, 54.9 – not estimable).

For newly diagnosed patients, the preset defined rate of interest of 55% for CCyR was reached as early as 6 months, and exceeded 90% by 12 and 24 months.

Dr Gore pointed out that intolerant patients also reached CCyR relatively quickly, although it was not a specified endpoint.

Data indicate that responses occurred relatively quickly and continued to increase over time of follow-up.

MMR also continued to increase over time and showed no difference between formulation and response rate.

Median PFS has not been reached, as only 7 patients in each cohort had disease progression.

One imatinib-R/I patient died 1 year after stopping treatment. The patient, who had a GI bleed unrelated to dasatinib, had discontinued therapy for progressive disease with loss of MCyR.

Safety

Overall safety was very similar to the dasatinib exposure and experience in adults, Dr Gore said, and there were no differences in events between PFOS and tablets.

One patient in the PFOS cohort had a dasatinib-related grade 3 hypersensitivity reaction, which resolved after discontinuation of dasatinib.

“What’s important here,” she said, “is that there were almost no adverse events of severity in either cohort, only 1 in the imatinib refractory and intolerant and 1 in the newly diagnosed cohorts.”

“Most importantly for those of us with a lot of experience in this field,” she added, “there were no occurrences of pleural effusion, pericardial effusion, pulmonary edema, pulmonary hypertension, or any vascular occlusive events in patients noted on this trial.”

“Additionally, for pediatricians, we care a lot about what happens to growth in these patients and prospectively we collected a lot of data related to growth parameters in bone growth and development."

Of the dasatinib-related adverse events occurring in 10% or more of patients, there were only 5 growth and development events noted out of the 113 patients treated and all were grade 1 or 2 events, Dr Gore pointed out.

In the R/I cohort, one patient had osteopenia and gynecomastia. At the time of data analysis, this event had resolved even though the patient continued on dasatinib.

“We believe our data suggests that dasatinib could be considered as a new standard of care for children with CML in chronic phase,” she said.

“It includes the advantage of a liquid formulation as well as the advantages of once daily dosing and administration without regard to fed or fasting state,” she added, “which for all of us who treat children know could be quite important.”

The study was funded by Bristol-Myers Squibb.

Child with cancer Photo by Bill Branson
Photo by Bill Branson
Child with cancer

CHICAGO—The largest ongoing and prospective trial of pediatric patients with chronic myeloid leukemia in chronic phase (CML-CP), according to the best knowledge of the investigators, has found dasatinib to be safe and effective as first- or second-line therapy for these children.

Patients refractory to or intolerant of imatinib had a major cytogenetic response (MCyR) by 3 months and responses at 12 and 24 months exceeded 90%.

Newly diagnosed patients had a complete cytogenetic response (CCyR) by 6 months.

“We believe our data suggests that dasatinib could be considered as a new standard of care (SOC) for children with CML in chronic phase,” said study author Lia Gore, MD, of the University of Colorado School of Medicine/Children’s Hospital Colorado in Aurora.

She presented the findings of the study at the ASCO 2017 Annual Meeting (abstract 10511).

Study design

CA 180-226 is a phase 2, open-label, nonrandomized, prospective study conducted in 18 countries. Patients younger than 18 years with newly diagnosed CML-CP, or imatinib-resistant/intolerant (R/I) CML-CP, or CML in accelerated phase, or Ph+ acute lymphoblastic leukemia (ALL) were enrolled on the study between March 2009 and September 2014.

Dr Gore’s presentation focused on the CML-CP patients in the study, both the newly diagnosed and the imatinib-R/I patients.

The imatinib-R/I patients received dasatinib 60 mg/m2 tablets once daily, and the newly diagnosed patients received the same tablet dosage daily or a powdered formulation for oral suspension (PFOS) of dasatinib at 72 mg/m2 daily.

Dr Gore noted the different dosage in the oral suspension formulation is based on bioavailability studies performed in adults, which was determined to be equivalent to the 60 mg/m2 tablet formulation.

Once accrual was reached in the tablet cohort, newly diagnosed patients were accrued to the PFOS cohort. The patients on PFOS could switch to tablets after a year or more on the oral suspension.

Patients remained on treatment until disease progression, unacceptable toxicity occurred, or the patient/physician preference.

All patients had a minimum follow-up of 2 years. The longest follow-up was more than 90 months.

Primary objectives of the study were MCyR greater than 30% for imatinib-R/I patients and complete CCyR greater than 55% for newly diagnosed patients.

Secondary objectives included time to and duration of response, major molecular response (MMR), progression-free survival (PFS), overall survival (OS), and safety.

Baseline patient characteristics

One hundred thirteen patients were treated across the 3 cohorts—29 in the imatinib-R/I receiving tablets, 51 newly diagnosed patients receiving tablets, and 33 newly diagnosed patients in the PFOS arm. A total of 84 patients had newly diagnosed disease.

Of the 29 imatinib-R/I patients, 25 were resistant, 2 intolerant, and 2 undetermined. And 6 of the 25 resistant patients had defined imatinib-resistance mutations.

Median age was 13.8 years in the imatinib-R/I cohort, 12.9 years in the newly diagnosed on tablets, and 11.7 years in the PFOS group. Other baseline characteristics were similar among the cohorts.

“Importantly, there were 3 patients less than 2 years of age, and a substantial proportion of patients were actually less than 12 years of age,” Dr Gore pointed out.

Dasatinib exposure

The median duration of therapy was 50 months and 42 months in the R/I and newly diagnosed cohorts, respectively.

Forty-eight percent of the imatinib-R/I patients and 73% of the newly diagnosed patients are still on treatment. A relatively small number of patients discontinued therapy.

The median duration of therapy was shorter in the PFOS cohort because they were enrolled only after accrual to the tablet cohort. However, they were also followed up for more than 2 years.

 

 

Of the 33 patients on PFOS, 22 eventually switched to tablet formulation.

Results

The primary endpoint for imatinib-R/I patients—MCyR greater than 30%—was reached by 3 months, and MCyR at 12 and 24 months exceeded 90%. The median time to response was 3.1 months (range, 2.8 – 4.1), and median duration of response was not yet reached (range, 54.9 – not estimable).

For newly diagnosed patients, the preset defined rate of interest of 55% for CCyR was reached as early as 6 months, and exceeded 90% by 12 and 24 months.

Dr Gore pointed out that intolerant patients also reached CCyR relatively quickly, although it was not a specified endpoint.

Data indicate that responses occurred relatively quickly and continued to increase over time of follow-up.

MMR also continued to increase over time and showed no difference between formulation and response rate.

Median PFS has not been reached, as only 7 patients in each cohort had disease progression.

One imatinib-R/I patient died 1 year after stopping treatment. The patient, who had a GI bleed unrelated to dasatinib, had discontinued therapy for progressive disease with loss of MCyR.

Safety

Overall safety was very similar to the dasatinib exposure and experience in adults, Dr Gore said, and there were no differences in events between PFOS and tablets.

One patient in the PFOS cohort had a dasatinib-related grade 3 hypersensitivity reaction, which resolved after discontinuation of dasatinib.

“What’s important here,” she said, “is that there were almost no adverse events of severity in either cohort, only 1 in the imatinib refractory and intolerant and 1 in the newly diagnosed cohorts.”

“Most importantly for those of us with a lot of experience in this field,” she added, “there were no occurrences of pleural effusion, pericardial effusion, pulmonary edema, pulmonary hypertension, or any vascular occlusive events in patients noted on this trial.”

“Additionally, for pediatricians, we care a lot about what happens to growth in these patients and prospectively we collected a lot of data related to growth parameters in bone growth and development."

Of the dasatinib-related adverse events occurring in 10% or more of patients, there were only 5 growth and development events noted out of the 113 patients treated and all were grade 1 or 2 events, Dr Gore pointed out.

In the R/I cohort, one patient had osteopenia and gynecomastia. At the time of data analysis, this event had resolved even though the patient continued on dasatinib.

“We believe our data suggests that dasatinib could be considered as a new standard of care for children with CML in chronic phase,” she said.

“It includes the advantage of a liquid formulation as well as the advantages of once daily dosing and administration without regard to fed or fasting state,” she added, “which for all of us who treat children know could be quite important.”

The study was funded by Bristol-Myers Squibb.

Publications
Publications
Topics
Article Type
Display Headline
Dasatinib potentially a new SOC for children with CML-CP
Display Headline
Dasatinib potentially a new SOC for children with CML-CP
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica