Chemo quadruples risk for myeloid cancers

Article Type
Changed
Fri, 01/04/2019 - 09:47
Display Headline
Chemo quadruples risk for myeloid cancers

ORLANDO – Patients who undergo cytotoxic chemotherapy, even in the modern era, are at increased risk for developing myeloid neoplasms, based on data from a cohort of nearly 750,000 adults who were initially treated with chemotherapy and survived at least 1 year after diagnosis.

In the cohort, the standardized incidence ratio (SIR) for treatment-related acute myeloid leukemia (tAML) or myelodysplastic syndrome (MDS) was four times greater than would be expected in the general population, reported Lindsay M. Morton, Ph.D., of the division of cancer epidemiology and genetics at the National Cancer Institute in Bethesda, Md.

RTEmagicC_6cd30bb_Morton_Lindsay_S_MD.jpg.jpg
Dr. Lindsay M. Morton

“We now demonstrate that there are elevated risks for treatment-related leukemia after treatment for a broad spectrum of first primary malignancies that are generally consistent with what we know about changing treatment practices,” she said at the American Society of Hematology annual meeting.

“The number of cancer survivors in the United States has increased dramatically, to reach nearly 14 million individuals today, and in just the next few years the number is expected to reach more than 18 million people, which means that the long-term health of this population is of great clinical importance as well as public health importance,” Dr. Morton emphasized.

The link between cytotoxic chemotherapy and leukemia risk has been known since the 1960s, with certain classes of agents carrying higher risks than others, including platinum-containing compounds, alkylating agents (for example, cyclophosphamide, melphalan, chlorambucil), topoisomerase II inhibitors (doxorubicin, daunorubicin, epirubicin, etc.), and antimetabolites (5-fluorauracil, capecitabine, gemcitabine, et al.).

Treatment-related leukemias are associated with higher doses of these agents, and the trend in contemporary medicine is to use more of these agents upfront for the treatment of primary malignancies. Yet estimates of the risk of tAML, MDS, or other malignancies have been hard to come by because of the relative rarity of cases and the sporadic reports in the literature, Dr. Morton said.

The investigators previously reported that risk for tAML and other myeloid neoplasms changed over time, and showed that since the 1990s there was an uptick in risk for patients treated with chemotherapy for cancers of bone, joints, and endometrium, and since 2000 for patients treated with chemotherapy for cancers of the esophagus, cervix and prostate.

For example, risks for tAML were higher in the 1970s for patients with ovarian cancer treated with melphalan, a highly leukemogenic agent, but dropped somewhat with the switch to platinum-based agents. Similarly, women with breast cancer had a comparatively high risk with the use of melphalan, a decline in risk with the introduction of cyclophosphamide, and then an increase with the addition of anthracyclines and dose-dense regimens.

Risk update

To get a better idea of the magnitude of risk in the modern era, Dr. Morton and colleagues sifted through Surveillance, Epidemiology, and End Results (SEER) data to identify a cohort of 746,007 adults who were initially treated with chemotherapy and survived for at least 1 year following a diagnosis with a first primary malignancy from 2000 through 2012. They calculated SIRs based on variables that included age, race, sex, malignancy type and treatment period.

They looked at four categories of myeloid neoplasms as defined by World Health Organization criteria: AML/MDS, chronic myeloid leukemia, myeloproliferative neoplasms (MPN) negative for BCR-ABL (Philadelphia-negative), and chronic myelomonocytic leukemia (CMML).

They found that 2,071 patients developed treatment-related AML/MDS, translating into a fourfold incidence compared with the general population (SIR 4.1, 95% confidence interval [CI] 3.9-4.2), 106 were diagnosed with CMML

They also identified novel risk for tAML/MDS after chemotherapy by malignancy (see table).

RTEmagicC_6cd30bb_115029_graphic.png.png

The investigators found that breast cancer, non-Hodgkin lymphoma, and lung cancer were most commonly associated with tAML/MDS (SIRs 4.1, 7.3, and 4.1, respectively, all significant).

In addition, although the overall numbers of cases were small, the investigators noted “strikingly elevated” risks for cancers of bone (SIR 35.1, CI. 16.9-64.6). testes (15.6, CI, 9.2-24.6), and soft tissue (12.6, CI=7.7-19.4),

Risk for tAML/MD was more modestly elevated for cancers of the brain, ovaries, endometrium, cervix, and prostate, and for Hodgkin lymphoma, chronic lymphocytic leukemia, and multiple myeloma.

Adding radiotherapy to chemotherapy for cancers of the breast, lung, and stomach cancers showed a trend toward heightened tAML/MDS risk, but this was not significant.

An elevated risk for CMML was also seen after chemotherapy for lung cancer (SIR 2.5, CI, 1.3-4.4), breast cancer (1.8, CI, 1.3-2.5), and non-Hodgkin lymphoma (2.1, CI, 1.2-3.4). There was elevated risk for CMML following chemotherapy for breast cancer (3.0, CI. 1.7-5.0) and non-Hodgkin lymphoma (4.2, CI, 2.4-6.9).

There were no increased risks for other myeloproliferative neoplasms after chemotherapy for any first primary cancer, however.

 

 

“This reminds us that with new uses of standard agents and introduction of new agents, it’s critical to carefully weigh the risks and benefits of systemic therapy,” Dr. Morton said.

The investigators plan to quantify risks associated with specific drugs and doses, she added.

The study was supported by the National Cancer Institute. Dr. Morton reported no relevant conflicts of interest to disclose.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – Patients who undergo cytotoxic chemotherapy, even in the modern era, are at increased risk for developing myeloid neoplasms, based on data from a cohort of nearly 750,000 adults who were initially treated with chemotherapy and survived at least 1 year after diagnosis.

In the cohort, the standardized incidence ratio (SIR) for treatment-related acute myeloid leukemia (tAML) or myelodysplastic syndrome (MDS) was four times greater than would be expected in the general population, reported Lindsay M. Morton, Ph.D., of the division of cancer epidemiology and genetics at the National Cancer Institute in Bethesda, Md.

RTEmagicC_6cd30bb_Morton_Lindsay_S_MD.jpg.jpg
Dr. Lindsay M. Morton

“We now demonstrate that there are elevated risks for treatment-related leukemia after treatment for a broad spectrum of first primary malignancies that are generally consistent with what we know about changing treatment practices,” she said at the American Society of Hematology annual meeting.

“The number of cancer survivors in the United States has increased dramatically, to reach nearly 14 million individuals today, and in just the next few years the number is expected to reach more than 18 million people, which means that the long-term health of this population is of great clinical importance as well as public health importance,” Dr. Morton emphasized.

The link between cytotoxic chemotherapy and leukemia risk has been known since the 1960s, with certain classes of agents carrying higher risks than others, including platinum-containing compounds, alkylating agents (for example, cyclophosphamide, melphalan, chlorambucil), topoisomerase II inhibitors (doxorubicin, daunorubicin, epirubicin, etc.), and antimetabolites (5-fluorauracil, capecitabine, gemcitabine, et al.).

Treatment-related leukemias are associated with higher doses of these agents, and the trend in contemporary medicine is to use more of these agents upfront for the treatment of primary malignancies. Yet estimates of the risk of tAML, MDS, or other malignancies have been hard to come by because of the relative rarity of cases and the sporadic reports in the literature, Dr. Morton said.

The investigators previously reported that risk for tAML and other myeloid neoplasms changed over time, and showed that since the 1990s there was an uptick in risk for patients treated with chemotherapy for cancers of bone, joints, and endometrium, and since 2000 for patients treated with chemotherapy for cancers of the esophagus, cervix and prostate.

For example, risks for tAML were higher in the 1970s for patients with ovarian cancer treated with melphalan, a highly leukemogenic agent, but dropped somewhat with the switch to platinum-based agents. Similarly, women with breast cancer had a comparatively high risk with the use of melphalan, a decline in risk with the introduction of cyclophosphamide, and then an increase with the addition of anthracyclines and dose-dense regimens.

Risk update

To get a better idea of the magnitude of risk in the modern era, Dr. Morton and colleagues sifted through Surveillance, Epidemiology, and End Results (SEER) data to identify a cohort of 746,007 adults who were initially treated with chemotherapy and survived for at least 1 year following a diagnosis with a first primary malignancy from 2000 through 2012. They calculated SIRs based on variables that included age, race, sex, malignancy type and treatment period.

They looked at four categories of myeloid neoplasms as defined by World Health Organization criteria: AML/MDS, chronic myeloid leukemia, myeloproliferative neoplasms (MPN) negative for BCR-ABL (Philadelphia-negative), and chronic myelomonocytic leukemia (CMML).

They found that 2,071 patients developed treatment-related AML/MDS, translating into a fourfold incidence compared with the general population (SIR 4.1, 95% confidence interval [CI] 3.9-4.2), 106 were diagnosed with CMML

They also identified novel risk for tAML/MDS after chemotherapy by malignancy (see table).

RTEmagicC_6cd30bb_115029_graphic.png.png

The investigators found that breast cancer, non-Hodgkin lymphoma, and lung cancer were most commonly associated with tAML/MDS (SIRs 4.1, 7.3, and 4.1, respectively, all significant).

In addition, although the overall numbers of cases were small, the investigators noted “strikingly elevated” risks for cancers of bone (SIR 35.1, CI. 16.9-64.6). testes (15.6, CI, 9.2-24.6), and soft tissue (12.6, CI=7.7-19.4),

Risk for tAML/MD was more modestly elevated for cancers of the brain, ovaries, endometrium, cervix, and prostate, and for Hodgkin lymphoma, chronic lymphocytic leukemia, and multiple myeloma.

Adding radiotherapy to chemotherapy for cancers of the breast, lung, and stomach cancers showed a trend toward heightened tAML/MDS risk, but this was not significant.

An elevated risk for CMML was also seen after chemotherapy for lung cancer (SIR 2.5, CI, 1.3-4.4), breast cancer (1.8, CI, 1.3-2.5), and non-Hodgkin lymphoma (2.1, CI, 1.2-3.4). There was elevated risk for CMML following chemotherapy for breast cancer (3.0, CI. 1.7-5.0) and non-Hodgkin lymphoma (4.2, CI, 2.4-6.9).

There were no increased risks for other myeloproliferative neoplasms after chemotherapy for any first primary cancer, however.

 

 

“This reminds us that with new uses of standard agents and introduction of new agents, it’s critical to carefully weigh the risks and benefits of systemic therapy,” Dr. Morton said.

The investigators plan to quantify risks associated with specific drugs and doses, she added.

The study was supported by the National Cancer Institute. Dr. Morton reported no relevant conflicts of interest to disclose.

ORLANDO – Patients who undergo cytotoxic chemotherapy, even in the modern era, are at increased risk for developing myeloid neoplasms, based on data from a cohort of nearly 750,000 adults who were initially treated with chemotherapy and survived at least 1 year after diagnosis.

In the cohort, the standardized incidence ratio (SIR) for treatment-related acute myeloid leukemia (tAML) or myelodysplastic syndrome (MDS) was four times greater than would be expected in the general population, reported Lindsay M. Morton, Ph.D., of the division of cancer epidemiology and genetics at the National Cancer Institute in Bethesda, Md.

RTEmagicC_6cd30bb_Morton_Lindsay_S_MD.jpg.jpg
Dr. Lindsay M. Morton

“We now demonstrate that there are elevated risks for treatment-related leukemia after treatment for a broad spectrum of first primary malignancies that are generally consistent with what we know about changing treatment practices,” she said at the American Society of Hematology annual meeting.

“The number of cancer survivors in the United States has increased dramatically, to reach nearly 14 million individuals today, and in just the next few years the number is expected to reach more than 18 million people, which means that the long-term health of this population is of great clinical importance as well as public health importance,” Dr. Morton emphasized.

The link between cytotoxic chemotherapy and leukemia risk has been known since the 1960s, with certain classes of agents carrying higher risks than others, including platinum-containing compounds, alkylating agents (for example, cyclophosphamide, melphalan, chlorambucil), topoisomerase II inhibitors (doxorubicin, daunorubicin, epirubicin, etc.), and antimetabolites (5-fluorauracil, capecitabine, gemcitabine, et al.).

Treatment-related leukemias are associated with higher doses of these agents, and the trend in contemporary medicine is to use more of these agents upfront for the treatment of primary malignancies. Yet estimates of the risk of tAML, MDS, or other malignancies have been hard to come by because of the relative rarity of cases and the sporadic reports in the literature, Dr. Morton said.

The investigators previously reported that risk for tAML and other myeloid neoplasms changed over time, and showed that since the 1990s there was an uptick in risk for patients treated with chemotherapy for cancers of bone, joints, and endometrium, and since 2000 for patients treated with chemotherapy for cancers of the esophagus, cervix and prostate.

For example, risks for tAML were higher in the 1970s for patients with ovarian cancer treated with melphalan, a highly leukemogenic agent, but dropped somewhat with the switch to platinum-based agents. Similarly, women with breast cancer had a comparatively high risk with the use of melphalan, a decline in risk with the introduction of cyclophosphamide, and then an increase with the addition of anthracyclines and dose-dense regimens.

Risk update

To get a better idea of the magnitude of risk in the modern era, Dr. Morton and colleagues sifted through Surveillance, Epidemiology, and End Results (SEER) data to identify a cohort of 746,007 adults who were initially treated with chemotherapy and survived for at least 1 year following a diagnosis with a first primary malignancy from 2000 through 2012. They calculated SIRs based on variables that included age, race, sex, malignancy type and treatment period.

They looked at four categories of myeloid neoplasms as defined by World Health Organization criteria: AML/MDS, chronic myeloid leukemia, myeloproliferative neoplasms (MPN) negative for BCR-ABL (Philadelphia-negative), and chronic myelomonocytic leukemia (CMML).

They found that 2,071 patients developed treatment-related AML/MDS, translating into a fourfold incidence compared with the general population (SIR 4.1, 95% confidence interval [CI] 3.9-4.2), 106 were diagnosed with CMML

They also identified novel risk for tAML/MDS after chemotherapy by malignancy (see table).

RTEmagicC_6cd30bb_115029_graphic.png.png

The investigators found that breast cancer, non-Hodgkin lymphoma, and lung cancer were most commonly associated with tAML/MDS (SIRs 4.1, 7.3, and 4.1, respectively, all significant).

In addition, although the overall numbers of cases were small, the investigators noted “strikingly elevated” risks for cancers of bone (SIR 35.1, CI. 16.9-64.6). testes (15.6, CI, 9.2-24.6), and soft tissue (12.6, CI=7.7-19.4),

Risk for tAML/MD was more modestly elevated for cancers of the brain, ovaries, endometrium, cervix, and prostate, and for Hodgkin lymphoma, chronic lymphocytic leukemia, and multiple myeloma.

Adding radiotherapy to chemotherapy for cancers of the breast, lung, and stomach cancers showed a trend toward heightened tAML/MDS risk, but this was not significant.

An elevated risk for CMML was also seen after chemotherapy for lung cancer (SIR 2.5, CI, 1.3-4.4), breast cancer (1.8, CI, 1.3-2.5), and non-Hodgkin lymphoma (2.1, CI, 1.2-3.4). There was elevated risk for CMML following chemotherapy for breast cancer (3.0, CI. 1.7-5.0) and non-Hodgkin lymphoma (4.2, CI, 2.4-6.9).

There were no increased risks for other myeloproliferative neoplasms after chemotherapy for any first primary cancer, however.

 

 

“This reminds us that with new uses of standard agents and introduction of new agents, it’s critical to carefully weigh the risks and benefits of systemic therapy,” Dr. Morton said.

The investigators plan to quantify risks associated with specific drugs and doses, she added.

The study was supported by the National Cancer Institute. Dr. Morton reported no relevant conflicts of interest to disclose.

References

References

Publications
Publications
Topics
Article Type
Display Headline
Chemo quadruples risk for myeloid cancers
Display Headline
Chemo quadruples risk for myeloid cancers
Sections
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

Vitals

<p><b>Key clinical point: </b>This study quantifies the risks for treatment-related myeloid cancers after chemotherapy.
</p><p><b>Major finding: </b>Chemotherapy is associated with a fourfold risk for treatment-related AML/MDS, compared with the general population.
</p><p><b>Data source: </b>Retrospective review of data on 746,007 adults treated with chemotherapy for a first primary malignancy.
</p><p><b>Disclosures: </b>The National Cancer Institute supported the study. Dr. Morton reported having no conflicts of interest to disclose.</p>

Anti-BCL2, CD20 combo safe, effective in untreated CLL

Article Type
Changed
Thu, 01/12/2023 - 10:46
Display Headline
Anti-BCL2, CD20 combo safe, effective in untreated CLL

ORLANDO – An experimental combination of obinutuzumab and venetoclax appears safe as frontline therapy for patients with active, untreated chronic lymphocytic leukemia and comorbidities.

In the safety run-in portion of a phase 3, open-label trial comparing the combination of obinutuzumab (Gazyva) and the investigational Bcl-2 inhibitor venetoclax with obinutuzumab and its usual partner chlorambucil in 12 patients, only two of seven patients classified as being at high risk for the tumor lysis syndrome (TLS) developed laboratory-defined TLS, and no patients had clinical TLS.

The combination did not meet any of the pre-specified stopping criteria, and early data hinted at efficacy for the combination, said Dr. Kirsten Fischer, from the Center for Integrated Oncology at University Hospital Cologne in Germany.

RTEmagicC_12a53ac_Fischer_Kirsten_GERMANY.jpg.jpg
Dr. Kirsten Fischer

“As with previous reports, our data confirm rapid and profound reduction in lymphocyte counts after the first dose obinutuzumab in all 12 [evaluable] patients,” she said at the American Society of Hematology annual meeting.

In the CLL 11 trial, investigators in the German CLL group previously showed that the combination of the anti-CD20 antibody obinutuzumab and chlorambucil resulted in improved overall survival compared to chlorambucil alone in patients with previously untreated CLL and coexisting medical conditions. The combination is approved in the United States for adults with treatment-naïve CLL.

Venetoclax has been shown to have good efficacy against relapsed/refractory CLL both as monotherapy and in combination with rituximab, prompting the investigators to explore it in combination with the rituximab follow-on drug obinutuzumab.

In the safety run-in phase of the CLL 14 trial, the investigators enrolled 13 adults (median age 75, range 59-88 years) with newly diagnosed, active confirmed CLL and coexisting medical conditions as determined either by a score greater 6 on the cumulative illness rating scale (CIRS) or by estimated creatinine clearance less than 70 mL/min.

The patients were treated with 6 cycles of obinutuzumab and venetoclax followed by 6 additional cycles of venetoclax. Obinutuzumab was administered intravenously 100 mg on day 1 and 900 mg on day 2, with the option to deliver 1,000 mg on day 1 instead, then 1,000 mg on days 8 and 15 of cycle 1, and 1,000 mg on day 1 for cycles 2-6.

The dose of venetoclax was titrated upward gradually, with doses of 20 mg, 50 mg, 100 mg, 200 mg, and up to 400 mg administered starting on day 22 of cycle 1.

Planned stopping criteria were one treatment-related death or a grade 4 adverse event related to clinical TLS despite prophylaxis as specified by the protocol.

At the time of data cutoff (October 2015), 12 patients had been on treatment for at least 4 weeks and had reached the maximum venetoclax dose. Two patients had reached 11 cycles, three had reached 10 cycles, and seven had reached 8 cycles.

Grade 1 or 2 adverse events in all 13 enrolled patients included infusion-related reactions in 8; infections in 6; diarrhea, hyperkalemia, and constipation in 5; nausea, dizziness and cough in 4; and fatigue, headache and pruritus in 3.

Grade 3 or 4 adverse events were neutropenia in five patients; infusion related reactions; syncope, thrombocytopenia and laboratory-defined TLS in two patients; and bradycardia, hyperglycemia, influenza, leucopenia, pyrexia, respiratory tract infection, and elevated transaminases in one patient each.

As noted, all 12 evaluable patients had rapid drops in absolute lymphocyte counts, and all but one had complete resolution of lymphadenopathy after three cycles, with the improvement maintained after six cycles. The remaining patient had a decrease to near normal after both three and six cycles.

Of the 12 patients, 11 had a partial response after three cycles, and the remaining patient had stable disease, for an overall response rate of 92%. The overall response rate after six cycles was 100%, with all patients having a partial response.

The data were sufficiently good to justify continuing with the randomized phase, which began in August 2015, Dr. Fischer noted.

The study is sponsored by Hoffman-La Roche and AbbVie. Dr. Fischer disclosed receiving travel grants from Hoffman-La Roche.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – An experimental combination of obinutuzumab and venetoclax appears safe as frontline therapy for patients with active, untreated chronic lymphocytic leukemia and comorbidities.

In the safety run-in portion of a phase 3, open-label trial comparing the combination of obinutuzumab (Gazyva) and the investigational Bcl-2 inhibitor venetoclax with obinutuzumab and its usual partner chlorambucil in 12 patients, only two of seven patients classified as being at high risk for the tumor lysis syndrome (TLS) developed laboratory-defined TLS, and no patients had clinical TLS.

The combination did not meet any of the pre-specified stopping criteria, and early data hinted at efficacy for the combination, said Dr. Kirsten Fischer, from the Center for Integrated Oncology at University Hospital Cologne in Germany.

RTEmagicC_12a53ac_Fischer_Kirsten_GERMANY.jpg.jpg
Dr. Kirsten Fischer

“As with previous reports, our data confirm rapid and profound reduction in lymphocyte counts after the first dose obinutuzumab in all 12 [evaluable] patients,” she said at the American Society of Hematology annual meeting.

In the CLL 11 trial, investigators in the German CLL group previously showed that the combination of the anti-CD20 antibody obinutuzumab and chlorambucil resulted in improved overall survival compared to chlorambucil alone in patients with previously untreated CLL and coexisting medical conditions. The combination is approved in the United States for adults with treatment-naïve CLL.

Venetoclax has been shown to have good efficacy against relapsed/refractory CLL both as monotherapy and in combination with rituximab, prompting the investigators to explore it in combination with the rituximab follow-on drug obinutuzumab.

In the safety run-in phase of the CLL 14 trial, the investigators enrolled 13 adults (median age 75, range 59-88 years) with newly diagnosed, active confirmed CLL and coexisting medical conditions as determined either by a score greater 6 on the cumulative illness rating scale (CIRS) or by estimated creatinine clearance less than 70 mL/min.

The patients were treated with 6 cycles of obinutuzumab and venetoclax followed by 6 additional cycles of venetoclax. Obinutuzumab was administered intravenously 100 mg on day 1 and 900 mg on day 2, with the option to deliver 1,000 mg on day 1 instead, then 1,000 mg on days 8 and 15 of cycle 1, and 1,000 mg on day 1 for cycles 2-6.

The dose of venetoclax was titrated upward gradually, with doses of 20 mg, 50 mg, 100 mg, 200 mg, and up to 400 mg administered starting on day 22 of cycle 1.

Planned stopping criteria were one treatment-related death or a grade 4 adverse event related to clinical TLS despite prophylaxis as specified by the protocol.

At the time of data cutoff (October 2015), 12 patients had been on treatment for at least 4 weeks and had reached the maximum venetoclax dose. Two patients had reached 11 cycles, three had reached 10 cycles, and seven had reached 8 cycles.

Grade 1 or 2 adverse events in all 13 enrolled patients included infusion-related reactions in 8; infections in 6; diarrhea, hyperkalemia, and constipation in 5; nausea, dizziness and cough in 4; and fatigue, headache and pruritus in 3.

Grade 3 or 4 adverse events were neutropenia in five patients; infusion related reactions; syncope, thrombocytopenia and laboratory-defined TLS in two patients; and bradycardia, hyperglycemia, influenza, leucopenia, pyrexia, respiratory tract infection, and elevated transaminases in one patient each.

As noted, all 12 evaluable patients had rapid drops in absolute lymphocyte counts, and all but one had complete resolution of lymphadenopathy after three cycles, with the improvement maintained after six cycles. The remaining patient had a decrease to near normal after both three and six cycles.

Of the 12 patients, 11 had a partial response after three cycles, and the remaining patient had stable disease, for an overall response rate of 92%. The overall response rate after six cycles was 100%, with all patients having a partial response.

The data were sufficiently good to justify continuing with the randomized phase, which began in August 2015, Dr. Fischer noted.

The study is sponsored by Hoffman-La Roche and AbbVie. Dr. Fischer disclosed receiving travel grants from Hoffman-La Roche.

ORLANDO – An experimental combination of obinutuzumab and venetoclax appears safe as frontline therapy for patients with active, untreated chronic lymphocytic leukemia and comorbidities.

In the safety run-in portion of a phase 3, open-label trial comparing the combination of obinutuzumab (Gazyva) and the investigational Bcl-2 inhibitor venetoclax with obinutuzumab and its usual partner chlorambucil in 12 patients, only two of seven patients classified as being at high risk for the tumor lysis syndrome (TLS) developed laboratory-defined TLS, and no patients had clinical TLS.

The combination did not meet any of the pre-specified stopping criteria, and early data hinted at efficacy for the combination, said Dr. Kirsten Fischer, from the Center for Integrated Oncology at University Hospital Cologne in Germany.

RTEmagicC_12a53ac_Fischer_Kirsten_GERMANY.jpg.jpg
Dr. Kirsten Fischer

“As with previous reports, our data confirm rapid and profound reduction in lymphocyte counts after the first dose obinutuzumab in all 12 [evaluable] patients,” she said at the American Society of Hematology annual meeting.

In the CLL 11 trial, investigators in the German CLL group previously showed that the combination of the anti-CD20 antibody obinutuzumab and chlorambucil resulted in improved overall survival compared to chlorambucil alone in patients with previously untreated CLL and coexisting medical conditions. The combination is approved in the United States for adults with treatment-naïve CLL.

Venetoclax has been shown to have good efficacy against relapsed/refractory CLL both as monotherapy and in combination with rituximab, prompting the investigators to explore it in combination with the rituximab follow-on drug obinutuzumab.

In the safety run-in phase of the CLL 14 trial, the investigators enrolled 13 adults (median age 75, range 59-88 years) with newly diagnosed, active confirmed CLL and coexisting medical conditions as determined either by a score greater 6 on the cumulative illness rating scale (CIRS) or by estimated creatinine clearance less than 70 mL/min.

The patients were treated with 6 cycles of obinutuzumab and venetoclax followed by 6 additional cycles of venetoclax. Obinutuzumab was administered intravenously 100 mg on day 1 and 900 mg on day 2, with the option to deliver 1,000 mg on day 1 instead, then 1,000 mg on days 8 and 15 of cycle 1, and 1,000 mg on day 1 for cycles 2-6.

The dose of venetoclax was titrated upward gradually, with doses of 20 mg, 50 mg, 100 mg, 200 mg, and up to 400 mg administered starting on day 22 of cycle 1.

Planned stopping criteria were one treatment-related death or a grade 4 adverse event related to clinical TLS despite prophylaxis as specified by the protocol.

At the time of data cutoff (October 2015), 12 patients had been on treatment for at least 4 weeks and had reached the maximum venetoclax dose. Two patients had reached 11 cycles, three had reached 10 cycles, and seven had reached 8 cycles.

Grade 1 or 2 adverse events in all 13 enrolled patients included infusion-related reactions in 8; infections in 6; diarrhea, hyperkalemia, and constipation in 5; nausea, dizziness and cough in 4; and fatigue, headache and pruritus in 3.

Grade 3 or 4 adverse events were neutropenia in five patients; infusion related reactions; syncope, thrombocytopenia and laboratory-defined TLS in two patients; and bradycardia, hyperglycemia, influenza, leucopenia, pyrexia, respiratory tract infection, and elevated transaminases in one patient each.

As noted, all 12 evaluable patients had rapid drops in absolute lymphocyte counts, and all but one had complete resolution of lymphadenopathy after three cycles, with the improvement maintained after six cycles. The remaining patient had a decrease to near normal after both three and six cycles.

Of the 12 patients, 11 had a partial response after three cycles, and the remaining patient had stable disease, for an overall response rate of 92%. The overall response rate after six cycles was 100%, with all patients having a partial response.

The data were sufficiently good to justify continuing with the randomized phase, which began in August 2015, Dr. Fischer noted.

The study is sponsored by Hoffman-La Roche and AbbVie. Dr. Fischer disclosed receiving travel grants from Hoffman-La Roche.

References

References

Publications
Publications
Topics
Article Type
Display Headline
Anti-BCL2, CD20 combo safe, effective in untreated CLL
Display Headline
Anti-BCL2, CD20 combo safe, effective in untreated CLL
Sections
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

Vitals

<p><b>Key clinical point:</b> Patients with CLL and comorbidities were able to tolerate an experimental regimen of obinutuzumab and venetoclax.
</p><p><b>Major finding:</b> Two of 12 evaluable patients had evidence of laboratory-defined but not clinical tumor lysis syndrome.
</p><p><b>Data source:</b> Safety run-in phase of a randomized phase 3 trial with 13 patients with chronic lymphocytic leukemia and comorbidities.
</p><p><b>Disclosures:</b> The study is sponsored by Hoffman-La Roche and AbbVie. Dr. Fischer disclosed receiving travel grants from Hoffman-La Roche.</p>

ASH: Idelalisib plus standard therapy boosts survival in relapsed CLL

Article Type
Changed
Thu, 01/12/2023 - 10:46
Display Headline
ASH: Idelalisib plus standard therapy boosts survival in relapsed CLL

ORLANDO – Adding the PI3K inhibitor idelalisib to a standard regimen of bendamustine and rituximab significantly reduced the risk of both disease progression and death for patients with relapsed and/or refractory chronic lymphocytic leukemia, results of a phase III randomized trial showed.

At a median follow-up of 12 months, the primary endpoint of median progression-free survival was 23.1 months for patients treated with idelalisib (Zydelig), bendamustine, and rituximab (idel+BR), compared with 11.1 months for bendamustine and rituximab (BR) plus a placebo, reported Dr. Andrew Zelenetz of Memorial Sloan Kettering Cancer Center, New York.

RTEmagicC_cd7690c_Zelenetz_Andrew_NYC.jpg.jpg
Dr. Andrew Zelenetz

“Median overall survival was not reached in either arm. However, there was a significant improvement in overall survival, with a 45% reduction in the risk of death [with idel+BR],” he said in a late-breaking abstract session at the annual meeting of the American Society of Hematology.

The trial was stopped early after a data review at the first planned interim analysis showed significant superiority for the three-drug combination.

The results were consistent across subgroups, including patients with high-risk features such as deletion 17p and mutated TP53 (del[17p]/TP53), unmutated immunoglobulin heavy chain variable region (IgHV), and treatment-refractory disease.

The rationale behind adding idelalisib, an inhibitor of the phosphatidylinositol-3 kinase (PI3K), is that signaling via the PI3K pathway is hyperactive and can be targeted, Dr. Zelenetz explained.

Study 115 was a phase III trial with accrual from June 2012 through August 2015. Investigators enrolled 416 patients with relapsed /refractory CLL and randomly assigned them to receive BR for six 28-day cycles of bendamustine (70 mg/m2 on days 1 and 2 of each cycle) and rituximab (375 mg/m2 for cycle 1, and 500 mg/m2 for cycles 2 through 6), plus either idelalisib 150 mg b.i.d. or placebo, each administered continuously until disease progression, intolerable toxicity, withdrawal of consent, or death.

The patients were stratified by mutational and disease status (refractory defined as CLL progression less than 6 months from completion of prior therapy, or relapsed CLL progression 6 months or more from completion of prior therapy.

The trial was halted early after the first planned interim analysis, which was conducted after 75% of the total number of 260 planned events of CLL progression or death from any cause had occurred. The data cutoff was June 15, 2015.

The intention-to-treat analysis included 207 patients assigned to idelalisib and 209 assigned to placebo. Three-fourths (76%) of the patients were male.

In all, 46% of patients had Rai stage III/IV disease. The median time since the completion of the last therapy was 16 months.

The proportions of patients with high-risk features included del(17p)/p53 mutation in 32.9%, unmutated IgHV in 83.2%, and treatment-refractory disease in 29.8%.

As noted, the median progression-free survival with idelalisib at a median follow-up of 12 months was 23.1 months vs. 11.1 months for placebo. That translated into a hazard ratio of 0.33 (P less than .0001).

Among patients with neither del(17p) nor TP53 mutations, the HR for progression was 0.22. Among patients with either del(17p) or a TP53 mutation, the HR was 0.50 (95% confidence intervals show statistical significance for both).

Overall response rates were 68% among patients who received idelalisib, and 45% for those who received placebo. There were five complete responses (2%) in the idelalisib group and none in the placebo group.

The idelalisib group also had a higher proportion of patients with a greater than 50% reduction in involved lymph nodes (96% vs. 61%), and had better organomegaly responses (spleen and liver) and hematologic responses (hemoglobin, neutrophils, and platelets).

Grade 3 or greater adverse events occurred in 93% of patients on idelalisib, compared with 76% of those on placebo. The proportion of patients with any serious adverse event was 66% vs. 44%, respectively.

Adverse events leading to drug dose reduction were seen in 11% of idelalisib-treated patients, compared with 6% of placebo controls, and therapy was discontinued in 26% vs. 13%, respectively.

Ten patients in the idelalisib arm and seven in the placebo arm died during the study.

Adverse events that occurred more commonly with idelalisib included neutropenia, pyrexia, diarrhea, febrile neutropenia, pneumonia, rash, and elevated liver enzymes.

Session moderator Dr. David P. Steensma of the Dana-Farber Cancer Institute in Boston asked Dr. Zelenetz how idelalisib plus BR stacked up to ibrutinib (Imbruvica) plus BR in this population.

Dr. Zelenetz noted that patients were excluded from the HELIOS trial of ibrutinib plus BR if they had del(17p). Comparing the subset of patients in Study 115 without del(17p) with patients in the ibrutinib study, “the results are virtually superimposable,” Dr. Zelenetz said, and “the two treatments are really remarkably similar.”

 

 

The overall survival benefit was larger in the HELIOS trial, Dr. Zelenetz noted, but that was largely because the trial allowed patients to cross over from placebo to the active drug.

Gilead Sciences funded Study 115. Dr. Zelenetz disclosed receiving research funding from the company and discussing off-label use of idelalisib for relapsed/refractory CLL.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
leukemia, chronic lymphocytic leukemia, idelalisib, CLL, bendamustine, rituximab
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – Adding the PI3K inhibitor idelalisib to a standard regimen of bendamustine and rituximab significantly reduced the risk of both disease progression and death for patients with relapsed and/or refractory chronic lymphocytic leukemia, results of a phase III randomized trial showed.

At a median follow-up of 12 months, the primary endpoint of median progression-free survival was 23.1 months for patients treated with idelalisib (Zydelig), bendamustine, and rituximab (idel+BR), compared with 11.1 months for bendamustine and rituximab (BR) plus a placebo, reported Dr. Andrew Zelenetz of Memorial Sloan Kettering Cancer Center, New York.

RTEmagicC_cd7690c_Zelenetz_Andrew_NYC.jpg.jpg
Dr. Andrew Zelenetz

“Median overall survival was not reached in either arm. However, there was a significant improvement in overall survival, with a 45% reduction in the risk of death [with idel+BR],” he said in a late-breaking abstract session at the annual meeting of the American Society of Hematology.

The trial was stopped early after a data review at the first planned interim analysis showed significant superiority for the three-drug combination.

The results were consistent across subgroups, including patients with high-risk features such as deletion 17p and mutated TP53 (del[17p]/TP53), unmutated immunoglobulin heavy chain variable region (IgHV), and treatment-refractory disease.

The rationale behind adding idelalisib, an inhibitor of the phosphatidylinositol-3 kinase (PI3K), is that signaling via the PI3K pathway is hyperactive and can be targeted, Dr. Zelenetz explained.

Study 115 was a phase III trial with accrual from June 2012 through August 2015. Investigators enrolled 416 patients with relapsed /refractory CLL and randomly assigned them to receive BR for six 28-day cycles of bendamustine (70 mg/m2 on days 1 and 2 of each cycle) and rituximab (375 mg/m2 for cycle 1, and 500 mg/m2 for cycles 2 through 6), plus either idelalisib 150 mg b.i.d. or placebo, each administered continuously until disease progression, intolerable toxicity, withdrawal of consent, or death.

The patients were stratified by mutational and disease status (refractory defined as CLL progression less than 6 months from completion of prior therapy, or relapsed CLL progression 6 months or more from completion of prior therapy.

The trial was halted early after the first planned interim analysis, which was conducted after 75% of the total number of 260 planned events of CLL progression or death from any cause had occurred. The data cutoff was June 15, 2015.

The intention-to-treat analysis included 207 patients assigned to idelalisib and 209 assigned to placebo. Three-fourths (76%) of the patients were male.

In all, 46% of patients had Rai stage III/IV disease. The median time since the completion of the last therapy was 16 months.

The proportions of patients with high-risk features included del(17p)/p53 mutation in 32.9%, unmutated IgHV in 83.2%, and treatment-refractory disease in 29.8%.

As noted, the median progression-free survival with idelalisib at a median follow-up of 12 months was 23.1 months vs. 11.1 months for placebo. That translated into a hazard ratio of 0.33 (P less than .0001).

Among patients with neither del(17p) nor TP53 mutations, the HR for progression was 0.22. Among patients with either del(17p) or a TP53 mutation, the HR was 0.50 (95% confidence intervals show statistical significance for both).

Overall response rates were 68% among patients who received idelalisib, and 45% for those who received placebo. There were five complete responses (2%) in the idelalisib group and none in the placebo group.

The idelalisib group also had a higher proportion of patients with a greater than 50% reduction in involved lymph nodes (96% vs. 61%), and had better organomegaly responses (spleen and liver) and hematologic responses (hemoglobin, neutrophils, and platelets).

Grade 3 or greater adverse events occurred in 93% of patients on idelalisib, compared with 76% of those on placebo. The proportion of patients with any serious adverse event was 66% vs. 44%, respectively.

Adverse events leading to drug dose reduction were seen in 11% of idelalisib-treated patients, compared with 6% of placebo controls, and therapy was discontinued in 26% vs. 13%, respectively.

Ten patients in the idelalisib arm and seven in the placebo arm died during the study.

Adverse events that occurred more commonly with idelalisib included neutropenia, pyrexia, diarrhea, febrile neutropenia, pneumonia, rash, and elevated liver enzymes.

Session moderator Dr. David P. Steensma of the Dana-Farber Cancer Institute in Boston asked Dr. Zelenetz how idelalisib plus BR stacked up to ibrutinib (Imbruvica) plus BR in this population.

Dr. Zelenetz noted that patients were excluded from the HELIOS trial of ibrutinib plus BR if they had del(17p). Comparing the subset of patients in Study 115 without del(17p) with patients in the ibrutinib study, “the results are virtually superimposable,” Dr. Zelenetz said, and “the two treatments are really remarkably similar.”

 

 

The overall survival benefit was larger in the HELIOS trial, Dr. Zelenetz noted, but that was largely because the trial allowed patients to cross over from placebo to the active drug.

Gilead Sciences funded Study 115. Dr. Zelenetz disclosed receiving research funding from the company and discussing off-label use of idelalisib for relapsed/refractory CLL.

ORLANDO – Adding the PI3K inhibitor idelalisib to a standard regimen of bendamustine and rituximab significantly reduced the risk of both disease progression and death for patients with relapsed and/or refractory chronic lymphocytic leukemia, results of a phase III randomized trial showed.

At a median follow-up of 12 months, the primary endpoint of median progression-free survival was 23.1 months for patients treated with idelalisib (Zydelig), bendamustine, and rituximab (idel+BR), compared with 11.1 months for bendamustine and rituximab (BR) plus a placebo, reported Dr. Andrew Zelenetz of Memorial Sloan Kettering Cancer Center, New York.

RTEmagicC_cd7690c_Zelenetz_Andrew_NYC.jpg.jpg
Dr. Andrew Zelenetz

“Median overall survival was not reached in either arm. However, there was a significant improvement in overall survival, with a 45% reduction in the risk of death [with idel+BR],” he said in a late-breaking abstract session at the annual meeting of the American Society of Hematology.

The trial was stopped early after a data review at the first planned interim analysis showed significant superiority for the three-drug combination.

The results were consistent across subgroups, including patients with high-risk features such as deletion 17p and mutated TP53 (del[17p]/TP53), unmutated immunoglobulin heavy chain variable region (IgHV), and treatment-refractory disease.

The rationale behind adding idelalisib, an inhibitor of the phosphatidylinositol-3 kinase (PI3K), is that signaling via the PI3K pathway is hyperactive and can be targeted, Dr. Zelenetz explained.

Study 115 was a phase III trial with accrual from June 2012 through August 2015. Investigators enrolled 416 patients with relapsed /refractory CLL and randomly assigned them to receive BR for six 28-day cycles of bendamustine (70 mg/m2 on days 1 and 2 of each cycle) and rituximab (375 mg/m2 for cycle 1, and 500 mg/m2 for cycles 2 through 6), plus either idelalisib 150 mg b.i.d. or placebo, each administered continuously until disease progression, intolerable toxicity, withdrawal of consent, or death.

The patients were stratified by mutational and disease status (refractory defined as CLL progression less than 6 months from completion of prior therapy, or relapsed CLL progression 6 months or more from completion of prior therapy.

The trial was halted early after the first planned interim analysis, which was conducted after 75% of the total number of 260 planned events of CLL progression or death from any cause had occurred. The data cutoff was June 15, 2015.

The intention-to-treat analysis included 207 patients assigned to idelalisib and 209 assigned to placebo. Three-fourths (76%) of the patients were male.

In all, 46% of patients had Rai stage III/IV disease. The median time since the completion of the last therapy was 16 months.

The proportions of patients with high-risk features included del(17p)/p53 mutation in 32.9%, unmutated IgHV in 83.2%, and treatment-refractory disease in 29.8%.

As noted, the median progression-free survival with idelalisib at a median follow-up of 12 months was 23.1 months vs. 11.1 months for placebo. That translated into a hazard ratio of 0.33 (P less than .0001).

Among patients with neither del(17p) nor TP53 mutations, the HR for progression was 0.22. Among patients with either del(17p) or a TP53 mutation, the HR was 0.50 (95% confidence intervals show statistical significance for both).

Overall response rates were 68% among patients who received idelalisib, and 45% for those who received placebo. There were five complete responses (2%) in the idelalisib group and none in the placebo group.

The idelalisib group also had a higher proportion of patients with a greater than 50% reduction in involved lymph nodes (96% vs. 61%), and had better organomegaly responses (spleen and liver) and hematologic responses (hemoglobin, neutrophils, and platelets).

Grade 3 or greater adverse events occurred in 93% of patients on idelalisib, compared with 76% of those on placebo. The proportion of patients with any serious adverse event was 66% vs. 44%, respectively.

Adverse events leading to drug dose reduction were seen in 11% of idelalisib-treated patients, compared with 6% of placebo controls, and therapy was discontinued in 26% vs. 13%, respectively.

Ten patients in the idelalisib arm and seven in the placebo arm died during the study.

Adverse events that occurred more commonly with idelalisib included neutropenia, pyrexia, diarrhea, febrile neutropenia, pneumonia, rash, and elevated liver enzymes.

Session moderator Dr. David P. Steensma of the Dana-Farber Cancer Institute in Boston asked Dr. Zelenetz how idelalisib plus BR stacked up to ibrutinib (Imbruvica) plus BR in this population.

Dr. Zelenetz noted that patients were excluded from the HELIOS trial of ibrutinib plus BR if they had del(17p). Comparing the subset of patients in Study 115 without del(17p) with patients in the ibrutinib study, “the results are virtually superimposable,” Dr. Zelenetz said, and “the two treatments are really remarkably similar.”

 

 

The overall survival benefit was larger in the HELIOS trial, Dr. Zelenetz noted, but that was largely because the trial allowed patients to cross over from placebo to the active drug.

Gilead Sciences funded Study 115. Dr. Zelenetz disclosed receiving research funding from the company and discussing off-label use of idelalisib for relapsed/refractory CLL.

References

References

Publications
Publications
Topics
Article Type
Display Headline
ASH: Idelalisib plus standard therapy boosts survival in relapsed CLL
Display Headline
ASH: Idelalisib plus standard therapy boosts survival in relapsed CLL
Legacy Keywords
leukemia, chronic lymphocytic leukemia, idelalisib, CLL, bendamustine, rituximab
Legacy Keywords
leukemia, chronic lymphocytic leukemia, idelalisib, CLL, bendamustine, rituximab
Sections
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

Vitals

<p><b>Key clinical point:</b> Adding the PI3K inhibitor idelalisib to bendamustine and rituximab significantly improved survival of patients with relapsed/refractory CLL.
</p><p><b>Major finding: </b>Median progression-free survival was 23.1 months for patients treated with idelalisib, bendamustine, and rituximab, compared with 11.1 months for bendamustine and rituximab plus placebo.
</p><p><b>Data source:</b> Randomized, controlled trial in 416 patients with relapsed/refractory CLL. The trial was halted early for superior efficacy with idelalisib.
</p><p><b>Disclosures: </b>Gilead Sciences funded Study 115. Dr. Zelenetz disclosed receiving research funding from the company and discussing off-label use of idelalisib for relapsed/refractory CLL.</p>

Upfront idelalisib carries high risk for acute liver toxicity

Article Type
Changed
Fri, 12/16/2022 - 11:38
Display Headline
Upfront idelalisib carries high risk for acute liver toxicity

ORLANDO – Idelalisib given as first-line therapy for patients with chronic lymphocytic leukemia carries a high risk of early fulminant hepatotoxicity requiring drug interruption and steroids, investigators reported.

Among 24 patients who received idelalisib (Zydelig) monotherapy in a phase II trial of a combination of idelalisib followed by idelalisib concurrent with ofatumumab (Arzerra) as first-line therapy for chronic lymphocytic leukemia (CLL), 12 patients developed acute hepatotoxicity, marked by rapidly soaring levels of transaminase within about 28 days of starting therapy. An additional four patients developed hepatotoxicity at around 130 days, noted Dr. Benjamin L. Lampson, a clinical fellow in medicine at the Dana-Farber Cancer Institute in Boston.

RTEmagicC_e6e846b_Lampson_Benjamin_BOSTON.jpg.jpg
Dr. Benjamin Lampson

“Multiple lines of evidence suggest that this early hepatotoxicity is immune mediated. The proportion of regulatory T cells in the peripheral blood decrease on idelalisib therapy, providing a possible explanation for the development of early hepatotoxicity,” he said at the annual meeting of the American Society of Hematology.

The toxicities occur more frequently among younger and less heavily pretreated patients, and are likely due to on-target, immune-mediated effects, he noted.

Dr. Lampson presented data on the first 24 patients in an ongoing phase II trial. Patients with previously untreated CLL receive idelalisib 150 mg twice daily for 56 days, in an attempt to mobilize neoplastic B cells from the peripheral lymphoid tissues and into the bloodstream.

Following the monotherapy phase, patients are given ofatumumab in an attempt to clear the disease from peripheral blood.

“This dosing strategy is slightly different than what has been previously been used in trials combining these particular drugs. Specifically, previously reported trials have started these agents simultaneously without a lead-in period of monotherapy,” Dr. Lampson explained.

When the lead-in phase is completed, patients receive idelalisib plus ofatumumab infusions once weekly for 8 weeks, followed by once-monthly infusions for 4 months. Patients then continue on idelalisib indefinitely. The primary endpoint is the overall response rate assessed 2 months after the completion of the combination therapy.

For the first 24 patients treated as of Nov. 9, 2015, the median time on therapy was 7.7 months and median follow-up was 14.7 months.

The median patient age was 67.4 years (range 57.6-84.9). CLL genetics showed that 13 patients had unmutated immunoglobulin heavy chain variable region (IgHV) disease, 4 had the 17p deletion and TP53 mutation, 1 had deletion 11q, and 13 had deletion 13q; some patients had more than one mutation.

“What we began to notice after enrolling just a few subjects on the trial was that severe hepatotoxicity was occurring shortly after initiating idelalisib,” Dr. Lampson said.

He presented one case, a 58-year-old man who was in the idelalisib monotherapy phase of the study. He developed grade 3 hepatotoxicity 28 days after starting the drug, despite having a normal liver function test just 1 day earlier. The drug was stopped, but his liver function tests continued to rise, suggesting a self-perpetuating or self-sustaining process.

On day 32, the patient was admitted to the hospital, and on day 33 he was started on steroids, based on the hypothesis that the hepatotoxicity might have been immune mediated. Two days after initiation of steroids, his liver function tests continued to rise, whereupon he was started on mycophenolate mofetil.

“With these two forms of immunosuppression, the [liver function tests] did eventually normalize, although the steroids and mycophenolate had to be tapered over a period of many weeks. And this patient was not the only patient to experience toxicity; in fact, hepatotoxicity was frequent and often severe,” he said.

At the time of maximum incidence, week 4, the percentage of patients with any hepatotoxicity was 46%, with 13% at grade 4, and 21% at grade 3.

“The median time to initial development of hepatotoxicity is 28 days. This suggests that the mechanism of hepatotoxicity is not immediate, but takes time to develop, consistent with an adaptive immune response. Furthermore, hepatotoxicity is typically occurring before the first dose of ofatumumab is occurring at week 8, suggesting idelalisib alone is the cause of the hepatotoxicity,” Dr. Lampson said.

A comparison of data from the ongoing study and from three previous studies – two with idelalisib in relapsed refractory disease, and one as first-line therapy in patients 65 and older – showed that grade 3 or greater hepatotoxicity was lowest in a phase I trial of idelalisib in which patients had received a median of five prior lines of therapy, occurring in only 1.9% of patients. In contrast, in the current study, 52% of patients experienced grade 3 or 4 transaminitis at some point in the trial.

 

 

Evidence for the hepatotoxicity being an on-target immune-mediated effect comes from lymphocytic infiltrate on liver biopsy and lymphocytic colitis in idelalisib-treated patients. Additional evidence comes from the fact that the toxicity is both treatable and preventable with steroids, he said.

He cautioned that hepatotoxicity can recur rapidly when the drug is reintroduced.

“In general, our experience has been if idelalisib is resumed while the subject remains on steroids, the drug is more likely to be tolerated and the subject eventually can be tapered off steroids,” he said.

Asked by an audience member whether patients who are receiving idelalisib in the first-line setting should also receive steroids, Dr. Lampson said that they closely monitor patient liver enzymes around 28 days, and if grade 1 transaminitis is detected, patients are automatically started on low-dose steroids.

The study is sponsored by the Dana-Farber Cancer Institute in collaboration with Gilead Sciences and GlaxoSmithKline. Dr. Lampson and colleagues declared no relevant conflicts of interest.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – Idelalisib given as first-line therapy for patients with chronic lymphocytic leukemia carries a high risk of early fulminant hepatotoxicity requiring drug interruption and steroids, investigators reported.

Among 24 patients who received idelalisib (Zydelig) monotherapy in a phase II trial of a combination of idelalisib followed by idelalisib concurrent with ofatumumab (Arzerra) as first-line therapy for chronic lymphocytic leukemia (CLL), 12 patients developed acute hepatotoxicity, marked by rapidly soaring levels of transaminase within about 28 days of starting therapy. An additional four patients developed hepatotoxicity at around 130 days, noted Dr. Benjamin L. Lampson, a clinical fellow in medicine at the Dana-Farber Cancer Institute in Boston.

RTEmagicC_e6e846b_Lampson_Benjamin_BOSTON.jpg.jpg
Dr. Benjamin Lampson

“Multiple lines of evidence suggest that this early hepatotoxicity is immune mediated. The proportion of regulatory T cells in the peripheral blood decrease on idelalisib therapy, providing a possible explanation for the development of early hepatotoxicity,” he said at the annual meeting of the American Society of Hematology.

The toxicities occur more frequently among younger and less heavily pretreated patients, and are likely due to on-target, immune-mediated effects, he noted.

Dr. Lampson presented data on the first 24 patients in an ongoing phase II trial. Patients with previously untreated CLL receive idelalisib 150 mg twice daily for 56 days, in an attempt to mobilize neoplastic B cells from the peripheral lymphoid tissues and into the bloodstream.

Following the monotherapy phase, patients are given ofatumumab in an attempt to clear the disease from peripheral blood.

“This dosing strategy is slightly different than what has been previously been used in trials combining these particular drugs. Specifically, previously reported trials have started these agents simultaneously without a lead-in period of monotherapy,” Dr. Lampson explained.

When the lead-in phase is completed, patients receive idelalisib plus ofatumumab infusions once weekly for 8 weeks, followed by once-monthly infusions for 4 months. Patients then continue on idelalisib indefinitely. The primary endpoint is the overall response rate assessed 2 months after the completion of the combination therapy.

For the first 24 patients treated as of Nov. 9, 2015, the median time on therapy was 7.7 months and median follow-up was 14.7 months.

The median patient age was 67.4 years (range 57.6-84.9). CLL genetics showed that 13 patients had unmutated immunoglobulin heavy chain variable region (IgHV) disease, 4 had the 17p deletion and TP53 mutation, 1 had deletion 11q, and 13 had deletion 13q; some patients had more than one mutation.

“What we began to notice after enrolling just a few subjects on the trial was that severe hepatotoxicity was occurring shortly after initiating idelalisib,” Dr. Lampson said.

He presented one case, a 58-year-old man who was in the idelalisib monotherapy phase of the study. He developed grade 3 hepatotoxicity 28 days after starting the drug, despite having a normal liver function test just 1 day earlier. The drug was stopped, but his liver function tests continued to rise, suggesting a self-perpetuating or self-sustaining process.

On day 32, the patient was admitted to the hospital, and on day 33 he was started on steroids, based on the hypothesis that the hepatotoxicity might have been immune mediated. Two days after initiation of steroids, his liver function tests continued to rise, whereupon he was started on mycophenolate mofetil.

“With these two forms of immunosuppression, the [liver function tests] did eventually normalize, although the steroids and mycophenolate had to be tapered over a period of many weeks. And this patient was not the only patient to experience toxicity; in fact, hepatotoxicity was frequent and often severe,” he said.

At the time of maximum incidence, week 4, the percentage of patients with any hepatotoxicity was 46%, with 13% at grade 4, and 21% at grade 3.

“The median time to initial development of hepatotoxicity is 28 days. This suggests that the mechanism of hepatotoxicity is not immediate, but takes time to develop, consistent with an adaptive immune response. Furthermore, hepatotoxicity is typically occurring before the first dose of ofatumumab is occurring at week 8, suggesting idelalisib alone is the cause of the hepatotoxicity,” Dr. Lampson said.

A comparison of data from the ongoing study and from three previous studies – two with idelalisib in relapsed refractory disease, and one as first-line therapy in patients 65 and older – showed that grade 3 or greater hepatotoxicity was lowest in a phase I trial of idelalisib in which patients had received a median of five prior lines of therapy, occurring in only 1.9% of patients. In contrast, in the current study, 52% of patients experienced grade 3 or 4 transaminitis at some point in the trial.

 

 

Evidence for the hepatotoxicity being an on-target immune-mediated effect comes from lymphocytic infiltrate on liver biopsy and lymphocytic colitis in idelalisib-treated patients. Additional evidence comes from the fact that the toxicity is both treatable and preventable with steroids, he said.

He cautioned that hepatotoxicity can recur rapidly when the drug is reintroduced.

“In general, our experience has been if idelalisib is resumed while the subject remains on steroids, the drug is more likely to be tolerated and the subject eventually can be tapered off steroids,” he said.

Asked by an audience member whether patients who are receiving idelalisib in the first-line setting should also receive steroids, Dr. Lampson said that they closely monitor patient liver enzymes around 28 days, and if grade 1 transaminitis is detected, patients are automatically started on low-dose steroids.

The study is sponsored by the Dana-Farber Cancer Institute in collaboration with Gilead Sciences and GlaxoSmithKline. Dr. Lampson and colleagues declared no relevant conflicts of interest.

ORLANDO – Idelalisib given as first-line therapy for patients with chronic lymphocytic leukemia carries a high risk of early fulminant hepatotoxicity requiring drug interruption and steroids, investigators reported.

Among 24 patients who received idelalisib (Zydelig) monotherapy in a phase II trial of a combination of idelalisib followed by idelalisib concurrent with ofatumumab (Arzerra) as first-line therapy for chronic lymphocytic leukemia (CLL), 12 patients developed acute hepatotoxicity, marked by rapidly soaring levels of transaminase within about 28 days of starting therapy. An additional four patients developed hepatotoxicity at around 130 days, noted Dr. Benjamin L. Lampson, a clinical fellow in medicine at the Dana-Farber Cancer Institute in Boston.

RTEmagicC_e6e846b_Lampson_Benjamin_BOSTON.jpg.jpg
Dr. Benjamin Lampson

“Multiple lines of evidence suggest that this early hepatotoxicity is immune mediated. The proportion of regulatory T cells in the peripheral blood decrease on idelalisib therapy, providing a possible explanation for the development of early hepatotoxicity,” he said at the annual meeting of the American Society of Hematology.

The toxicities occur more frequently among younger and less heavily pretreated patients, and are likely due to on-target, immune-mediated effects, he noted.

Dr. Lampson presented data on the first 24 patients in an ongoing phase II trial. Patients with previously untreated CLL receive idelalisib 150 mg twice daily for 56 days, in an attempt to mobilize neoplastic B cells from the peripheral lymphoid tissues and into the bloodstream.

Following the monotherapy phase, patients are given ofatumumab in an attempt to clear the disease from peripheral blood.

“This dosing strategy is slightly different than what has been previously been used in trials combining these particular drugs. Specifically, previously reported trials have started these agents simultaneously without a lead-in period of monotherapy,” Dr. Lampson explained.

When the lead-in phase is completed, patients receive idelalisib plus ofatumumab infusions once weekly for 8 weeks, followed by once-monthly infusions for 4 months. Patients then continue on idelalisib indefinitely. The primary endpoint is the overall response rate assessed 2 months after the completion of the combination therapy.

For the first 24 patients treated as of Nov. 9, 2015, the median time on therapy was 7.7 months and median follow-up was 14.7 months.

The median patient age was 67.4 years (range 57.6-84.9). CLL genetics showed that 13 patients had unmutated immunoglobulin heavy chain variable region (IgHV) disease, 4 had the 17p deletion and TP53 mutation, 1 had deletion 11q, and 13 had deletion 13q; some patients had more than one mutation.

“What we began to notice after enrolling just a few subjects on the trial was that severe hepatotoxicity was occurring shortly after initiating idelalisib,” Dr. Lampson said.

He presented one case, a 58-year-old man who was in the idelalisib monotherapy phase of the study. He developed grade 3 hepatotoxicity 28 days after starting the drug, despite having a normal liver function test just 1 day earlier. The drug was stopped, but his liver function tests continued to rise, suggesting a self-perpetuating or self-sustaining process.

On day 32, the patient was admitted to the hospital, and on day 33 he was started on steroids, based on the hypothesis that the hepatotoxicity might have been immune mediated. Two days after initiation of steroids, his liver function tests continued to rise, whereupon he was started on mycophenolate mofetil.

“With these two forms of immunosuppression, the [liver function tests] did eventually normalize, although the steroids and mycophenolate had to be tapered over a period of many weeks. And this patient was not the only patient to experience toxicity; in fact, hepatotoxicity was frequent and often severe,” he said.

At the time of maximum incidence, week 4, the percentage of patients with any hepatotoxicity was 46%, with 13% at grade 4, and 21% at grade 3.

“The median time to initial development of hepatotoxicity is 28 days. This suggests that the mechanism of hepatotoxicity is not immediate, but takes time to develop, consistent with an adaptive immune response. Furthermore, hepatotoxicity is typically occurring before the first dose of ofatumumab is occurring at week 8, suggesting idelalisib alone is the cause of the hepatotoxicity,” Dr. Lampson said.

A comparison of data from the ongoing study and from three previous studies – two with idelalisib in relapsed refractory disease, and one as first-line therapy in patients 65 and older – showed that grade 3 or greater hepatotoxicity was lowest in a phase I trial of idelalisib in which patients had received a median of five prior lines of therapy, occurring in only 1.9% of patients. In contrast, in the current study, 52% of patients experienced grade 3 or 4 transaminitis at some point in the trial.

 

 

Evidence for the hepatotoxicity being an on-target immune-mediated effect comes from lymphocytic infiltrate on liver biopsy and lymphocytic colitis in idelalisib-treated patients. Additional evidence comes from the fact that the toxicity is both treatable and preventable with steroids, he said.

He cautioned that hepatotoxicity can recur rapidly when the drug is reintroduced.

“In general, our experience has been if idelalisib is resumed while the subject remains on steroids, the drug is more likely to be tolerated and the subject eventually can be tapered off steroids,” he said.

Asked by an audience member whether patients who are receiving idelalisib in the first-line setting should also receive steroids, Dr. Lampson said that they closely monitor patient liver enzymes around 28 days, and if grade 1 transaminitis is detected, patients are automatically started on low-dose steroids.

The study is sponsored by the Dana-Farber Cancer Institute in collaboration with Gilead Sciences and GlaxoSmithKline. Dr. Lampson and colleagues declared no relevant conflicts of interest.

References

References

Publications
Publications
Topics
Article Type
Display Headline
Upfront idelalisib carries high risk for acute liver toxicity
Display Headline
Upfront idelalisib carries high risk for acute liver toxicity
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

Vitals

<p><b>Key clinical point: </b>Idelalisib in the first-line setting is associated with significant risk of hepatotoxicity, with a peak incidence at about 28 days of therapy.
</p><p><b>Major finding: </b>More than half (52%) of patients with newly diagnosed chronic lymphocytic leukemia had grade 3 or 4 hepatotoxicity with idelalisib monotherapy.
</p><p><b>Data source: </b>Ongoing phase II clinical trial with data on 24 patients.
</p><p><b>Disclosures: </b>The study is sponsored by the Dana-Farber Cancer Institute in collaboration with Gilead Sciences and GlaxoSmithKline. Dr. Lampson and colleagues declared no relevant conflicts of interest.</p>

VIDEO: Novel GBT440 improves blood parameters in sickle cell disease

Article Type
Changed
Fri, 01/18/2019 - 15:30
Display Headline
VIDEO: Novel GBT440 improves blood parameters in sickle cell disease

ORLANDO – A novel small molecule agent improved hematologic parameters and was associated with significant reduction in sickling of red blood cells in patients with sickle cell disease.

The drug was shown to increase hemoglobin in both healthy volunteers and patients, reduce reticulocytosis, and improve biomarkers of hemolysis and inflammation.

In an interview, Dr. Claire Hemmaway of Queens Hospital in Romford, England, discusses early results from a phase I/II randomized, double-blind, placebo-controlled, parallel-group trial.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
sickle cell disease
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – A novel small molecule agent improved hematologic parameters and was associated with significant reduction in sickling of red blood cells in patients with sickle cell disease.

The drug was shown to increase hemoglobin in both healthy volunteers and patients, reduce reticulocytosis, and improve biomarkers of hemolysis and inflammation.

In an interview, Dr. Claire Hemmaway of Queens Hospital in Romford, England, discusses early results from a phase I/II randomized, double-blind, placebo-controlled, parallel-group trial.

ORLANDO – A novel small molecule agent improved hematologic parameters and was associated with significant reduction in sickling of red blood cells in patients with sickle cell disease.

The drug was shown to increase hemoglobin in both healthy volunteers and patients, reduce reticulocytosis, and improve biomarkers of hemolysis and inflammation.

In an interview, Dr. Claire Hemmaway of Queens Hospital in Romford, England, discusses early results from a phase I/II randomized, double-blind, placebo-controlled, parallel-group trial.

References

References

Publications
Publications
Topics
Article Type
Display Headline
VIDEO: Novel GBT440 improves blood parameters in sickle cell disease
Display Headline
VIDEO: Novel GBT440 improves blood parameters in sickle cell disease
Legacy Keywords
sickle cell disease
Legacy Keywords
sickle cell disease
Sections
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

ASH: Daratumumab and len/dex produce high response rates in refractory myeloma

Article Type
Changed
Fri, 01/18/2019 - 15:30
Display Headline
ASH: Daratumumab and len/dex produce high response rates in refractory myeloma

ORLANDO – A combination of two standard therapies and the newly approved targeted agent daratumumab was associated with a high overall response rate and induced rapid, deep, and durable responses in patients with relapsed or refractory multiple myeloma, according to results of a phase I/II study presented at the annual meeting of the American Society of Hematology.

The median time to first response was 1 month; median time to best response was 5.1 months; and the median duration of response had not been reached at the most recent analysis. At 1 year, 91% of 26 patients with a partial response or better had not experienced disease progression. The 18-month progression-free survival rate was 72%, and the 18-month overall survival rate was 90%.

RTEmagicC_7385761_Plesner_Torben_DENMARK.jpg.jpg
Dr. Torben Plesner

“I have been looking forward to this day since I dosed the first patient with daratumumab in 2007. We have come a long way,” Dr. Torben Plesner said at a briefing at the meeting.

The study is one of several trials examining the use of daratumumab (Darzalex) in various combination regimens. Daratumumab (Darzalex) was recently approved by the Food and Drug Administration as monotherapy for use in patients with relapsed or refractory multiple myeloma for whom three or more prior lines of therapy, including proteasome inhibitors or immunomodulators, have failed.

Updated data on an expansion cohort from the study show that the combination of lenalidomide and dexamethasone (len/dex) with daratumumab was associated with an 81% (26 patients) overall response rate (ORR). This included stringent complete responses (sCR) in 25% (8 patients), complete responses in 9% (3 patients), and very good partial responses in 28% (9 patients), and 6 partial responses

“The responses were durable and they occurred rapidly,” said Dr. Plesner of Vejle Hospital and the University of Southern Denmark in Vejle.

Dr. Plesner presented updated results from the expansion cohort of the GEN503 study looking at the combination of daratumumab and len/dex.

In the open-label, single-arm study, patients received 16 mg/kg intravenous infusions of daratumumab once weekly for the first 2 months, twice weekly for months 3 through 6, and once every 4 weeks thereafter, plus oral lenalidomide 25 mg on days 1 through 21 of every 28-day cycle, and oral dexamethasone 40 mg once weekly.

In the dose-escalation phase, patients with relapsed multiple myeloma following two to four prior lines of therapy were enrolled. In the expansion cohort phase, patients with relapsed disease after at least one line of therapy could be enrolled, with no upper limit on the number of prior regimens.

The primary endpoint was adverse events; the most common were neutropenia in 84% of patients, cough in 50%, diarrhea, and muscle spasms (44% each).

Half of all patients had a serious adverse event, but the only events that occurred in more than one patient were neutropenia (three), gastroenteritis (two), and pyrexia (two). Infusion-related reactions, primarily during the first infusion, occurred in 56%, and the severity was grade 2 or less. Reactions were managed either with premedication or by slowing the infusion rate.

Two randomized phase III studies of the combination of daratumumab and len/dex are ongoing: the POLLUX trial, looking at the drugs in patients with relapsed/refractory disease, and the MAIA trial, using the combination as first-line therapy for newly diagnosed patients.

Speaking about the wealth of new therapies for multiple myeloma and other hematologic malignancies discussed at the briefing, Dr. Robert Hromas, professor of medicine at the University of Florida in Gainesville, commented that “I’ve been in the field for years and I can’t explain the excitement among blood doctors. It’s extraordinary, but we see plateaus lasting years now.”

“It’s so much fun to be a blood doctor,” he added.

The GEN503 trial is sponsored by Janssen. Dr. Plesner disclosed serving on an advisory board for, and receiving research funding from, Janssen. Dr. Hromas had no relevant disclosures.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – A combination of two standard therapies and the newly approved targeted agent daratumumab was associated with a high overall response rate and induced rapid, deep, and durable responses in patients with relapsed or refractory multiple myeloma, according to results of a phase I/II study presented at the annual meeting of the American Society of Hematology.

The median time to first response was 1 month; median time to best response was 5.1 months; and the median duration of response had not been reached at the most recent analysis. At 1 year, 91% of 26 patients with a partial response or better had not experienced disease progression. The 18-month progression-free survival rate was 72%, and the 18-month overall survival rate was 90%.

RTEmagicC_7385761_Plesner_Torben_DENMARK.jpg.jpg
Dr. Torben Plesner

“I have been looking forward to this day since I dosed the first patient with daratumumab in 2007. We have come a long way,” Dr. Torben Plesner said at a briefing at the meeting.

The study is one of several trials examining the use of daratumumab (Darzalex) in various combination regimens. Daratumumab (Darzalex) was recently approved by the Food and Drug Administration as monotherapy for use in patients with relapsed or refractory multiple myeloma for whom three or more prior lines of therapy, including proteasome inhibitors or immunomodulators, have failed.

Updated data on an expansion cohort from the study show that the combination of lenalidomide and dexamethasone (len/dex) with daratumumab was associated with an 81% (26 patients) overall response rate (ORR). This included stringent complete responses (sCR) in 25% (8 patients), complete responses in 9% (3 patients), and very good partial responses in 28% (9 patients), and 6 partial responses

“The responses were durable and they occurred rapidly,” said Dr. Plesner of Vejle Hospital and the University of Southern Denmark in Vejle.

Dr. Plesner presented updated results from the expansion cohort of the GEN503 study looking at the combination of daratumumab and len/dex.

In the open-label, single-arm study, patients received 16 mg/kg intravenous infusions of daratumumab once weekly for the first 2 months, twice weekly for months 3 through 6, and once every 4 weeks thereafter, plus oral lenalidomide 25 mg on days 1 through 21 of every 28-day cycle, and oral dexamethasone 40 mg once weekly.

In the dose-escalation phase, patients with relapsed multiple myeloma following two to four prior lines of therapy were enrolled. In the expansion cohort phase, patients with relapsed disease after at least one line of therapy could be enrolled, with no upper limit on the number of prior regimens.

The primary endpoint was adverse events; the most common were neutropenia in 84% of patients, cough in 50%, diarrhea, and muscle spasms (44% each).

Half of all patients had a serious adverse event, but the only events that occurred in more than one patient were neutropenia (three), gastroenteritis (two), and pyrexia (two). Infusion-related reactions, primarily during the first infusion, occurred in 56%, and the severity was grade 2 or less. Reactions were managed either with premedication or by slowing the infusion rate.

Two randomized phase III studies of the combination of daratumumab and len/dex are ongoing: the POLLUX trial, looking at the drugs in patients with relapsed/refractory disease, and the MAIA trial, using the combination as first-line therapy for newly diagnosed patients.

Speaking about the wealth of new therapies for multiple myeloma and other hematologic malignancies discussed at the briefing, Dr. Robert Hromas, professor of medicine at the University of Florida in Gainesville, commented that “I’ve been in the field for years and I can’t explain the excitement among blood doctors. It’s extraordinary, but we see plateaus lasting years now.”

“It’s so much fun to be a blood doctor,” he added.

The GEN503 trial is sponsored by Janssen. Dr. Plesner disclosed serving on an advisory board for, and receiving research funding from, Janssen. Dr. Hromas had no relevant disclosures.

ORLANDO – A combination of two standard therapies and the newly approved targeted agent daratumumab was associated with a high overall response rate and induced rapid, deep, and durable responses in patients with relapsed or refractory multiple myeloma, according to results of a phase I/II study presented at the annual meeting of the American Society of Hematology.

The median time to first response was 1 month; median time to best response was 5.1 months; and the median duration of response had not been reached at the most recent analysis. At 1 year, 91% of 26 patients with a partial response or better had not experienced disease progression. The 18-month progression-free survival rate was 72%, and the 18-month overall survival rate was 90%.

RTEmagicC_7385761_Plesner_Torben_DENMARK.jpg.jpg
Dr. Torben Plesner

“I have been looking forward to this day since I dosed the first patient with daratumumab in 2007. We have come a long way,” Dr. Torben Plesner said at a briefing at the meeting.

The study is one of several trials examining the use of daratumumab (Darzalex) in various combination regimens. Daratumumab (Darzalex) was recently approved by the Food and Drug Administration as monotherapy for use in patients with relapsed or refractory multiple myeloma for whom three or more prior lines of therapy, including proteasome inhibitors or immunomodulators, have failed.

Updated data on an expansion cohort from the study show that the combination of lenalidomide and dexamethasone (len/dex) with daratumumab was associated with an 81% (26 patients) overall response rate (ORR). This included stringent complete responses (sCR) in 25% (8 patients), complete responses in 9% (3 patients), and very good partial responses in 28% (9 patients), and 6 partial responses

“The responses were durable and they occurred rapidly,” said Dr. Plesner of Vejle Hospital and the University of Southern Denmark in Vejle.

Dr. Plesner presented updated results from the expansion cohort of the GEN503 study looking at the combination of daratumumab and len/dex.

In the open-label, single-arm study, patients received 16 mg/kg intravenous infusions of daratumumab once weekly for the first 2 months, twice weekly for months 3 through 6, and once every 4 weeks thereafter, plus oral lenalidomide 25 mg on days 1 through 21 of every 28-day cycle, and oral dexamethasone 40 mg once weekly.

In the dose-escalation phase, patients with relapsed multiple myeloma following two to four prior lines of therapy were enrolled. In the expansion cohort phase, patients with relapsed disease after at least one line of therapy could be enrolled, with no upper limit on the number of prior regimens.

The primary endpoint was adverse events; the most common were neutropenia in 84% of patients, cough in 50%, diarrhea, and muscle spasms (44% each).

Half of all patients had a serious adverse event, but the only events that occurred in more than one patient were neutropenia (three), gastroenteritis (two), and pyrexia (two). Infusion-related reactions, primarily during the first infusion, occurred in 56%, and the severity was grade 2 or less. Reactions were managed either with premedication or by slowing the infusion rate.

Two randomized phase III studies of the combination of daratumumab and len/dex are ongoing: the POLLUX trial, looking at the drugs in patients with relapsed/refractory disease, and the MAIA trial, using the combination as first-line therapy for newly diagnosed patients.

Speaking about the wealth of new therapies for multiple myeloma and other hematologic malignancies discussed at the briefing, Dr. Robert Hromas, professor of medicine at the University of Florida in Gainesville, commented that “I’ve been in the field for years and I can’t explain the excitement among blood doctors. It’s extraordinary, but we see plateaus lasting years now.”

“It’s so much fun to be a blood doctor,” he added.

The GEN503 trial is sponsored by Janssen. Dr. Plesner disclosed serving on an advisory board for, and receiving research funding from, Janssen. Dr. Hromas had no relevant disclosures.

References

References

Publications
Publications
Topics
Article Type
Display Headline
ASH: Daratumumab and len/dex produce high response rates in refractory myeloma
Display Headline
ASH: Daratumumab and len/dex produce high response rates in refractory myeloma
Sections
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

Vitals

<p><b>Key clinical point:</b> The combination of the targeted agent daratumumab (Darzalex) with len/dex was highly effective in heavily pretreated patients with multiple myeloma.
</p><p><b>Major finding:</b> The overall response rate was 81% (26 of 32 patients).
</p><p><b>Data source:</b> Open-label, single-arm expansion cohort from a phase I/II trial in 32 patients.
</p><p><b>Disclosures: </b>The GEN503 trial is sponsored by Janssen. Dr. Plesner disclosed serving on an advisory board for, and receiving research funding from, Janssen.</p>

ASH: All-oral regimen extends multiple myeloma PFS

Article Type
Changed
Fri, 01/04/2019 - 09:46
Display Headline
ASH: All-oral regimen extends multiple myeloma PFS

ORLANDO – The first all-oral drug combination for treatment of relapsed/refractory multiple myeloma significantly extended progression-free survival (PFS), based on the first of three planned interim analyses from a phase 3 trial.

The combination of the oral proteasome inhibitor ixazomib (Ninlaro, recently approved by the U.S. Food and Drug Administration) with lenalidomide and dexamethasone (len/dex) was associated with a 35% improvement in PFS, compared with len/dex and placebo, reported Dr. Philippe Moreau from Université Hospital of Nantes (France), Hôtel-Dieu.

The ixazomib combination also was associated with a significantly better median time to progression, at 21.4 vs. 15.7 months (P = .007). The time to response and duration of response were not significantly different, however, for the two groups in the placebo-controlled, randomized study of 722 patients.

“Ixazomib, when combined with len/dex for patients with relapsed and/or refractory multiple myeloma, was associated a significant and meaningful improvement in progression-free survival, significantly improved time to progression, and confirmed response rates,” Dr. Moreau said at a briefing at the American Society of Hematology annual meeting.

Dr. Moreau presented results of the phase III Tourmaline study, which compared len/dex plus weekly ixazomib or len/dex with weekly placebo in adults who have relapsed/refractory multiple myeloma following one to three prior lines of therapy and were not refractory to either prior lenalidomide or proteasome inhibitor–based therapy.

The study met its primary endpoint of a PFS advantage for the addition of ixazomib. The median PFS for ixazomib plus len/dex was 20.6 months, compared with 14.7 months for len/dex plus placebo. The hazard ratio for PFS with ixazomib-containing combination was 0.742 (P = .012).

The confirmed overall response rate (partial response or greater) was 78% with ixazomib vs. 71% with placebo (P = .035). The respective very good partial response or better rates were 48% vs. 39% (P = .014).

A total of 722 patients were randomized to either ixazomib 4 mg or placebo weekly on days 1, 8, 15 and 22 of each 28-day cycle, with oral lenalidomide 25 mg on days 1 through 21 (with dose reductions in patients with renal impairment at the investigator’s discretion) and oral dexamethasone 40 mg on days 1, 8, 15, and 22.

Patients were stratified at randomization by the number of prior therapies (one vs. two or three), previous proteasome-inhibitor exposure, and by International Staging System for multiple myeloma stage I or II vs. III. Cycles were repeated until disease progression or until patients experienced unacceptable toxicity.

The overall median age of patients was 66 (range, 30-91) years. In all, 70% had received a prior proteasome inhibitor, 88% had International Staging System stage I or II disease, and 59% had received just one prior line of therapy.

Dr. Moreau noted that the responses were rapid and durable with a median time to response of 1.1 months for ixazomib and 1.9 months for placebo, and a median duration of response of 20.5 months and 15 months, respectively. Neither difference was significant.

The incidence of grade 3 or greater adverse events was 68% for patients on ixazomib vs. 61% with placebo. A higher incidence of thrombocytopenia with ixazomib (19% vs. 9%) primarily accounted for the difference.

Dr. Moreau noted that peripheral neuropathy, a common problem with intravenous proteasome inhibitors, was low with ixazomib, and patients did not report significant changes in their quality of life.

The study was sponsored by Millennium Pharmaceuticals. Dr. Moreau disclosed receiving honoraria from the company.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – The first all-oral drug combination for treatment of relapsed/refractory multiple myeloma significantly extended progression-free survival (PFS), based on the first of three planned interim analyses from a phase 3 trial.

The combination of the oral proteasome inhibitor ixazomib (Ninlaro, recently approved by the U.S. Food and Drug Administration) with lenalidomide and dexamethasone (len/dex) was associated with a 35% improvement in PFS, compared with len/dex and placebo, reported Dr. Philippe Moreau from Université Hospital of Nantes (France), Hôtel-Dieu.

The ixazomib combination also was associated with a significantly better median time to progression, at 21.4 vs. 15.7 months (P = .007). The time to response and duration of response were not significantly different, however, for the two groups in the placebo-controlled, randomized study of 722 patients.

“Ixazomib, when combined with len/dex for patients with relapsed and/or refractory multiple myeloma, was associated a significant and meaningful improvement in progression-free survival, significantly improved time to progression, and confirmed response rates,” Dr. Moreau said at a briefing at the American Society of Hematology annual meeting.

Dr. Moreau presented results of the phase III Tourmaline study, which compared len/dex plus weekly ixazomib or len/dex with weekly placebo in adults who have relapsed/refractory multiple myeloma following one to three prior lines of therapy and were not refractory to either prior lenalidomide or proteasome inhibitor–based therapy.

The study met its primary endpoint of a PFS advantage for the addition of ixazomib. The median PFS for ixazomib plus len/dex was 20.6 months, compared with 14.7 months for len/dex plus placebo. The hazard ratio for PFS with ixazomib-containing combination was 0.742 (P = .012).

The confirmed overall response rate (partial response or greater) was 78% with ixazomib vs. 71% with placebo (P = .035). The respective very good partial response or better rates were 48% vs. 39% (P = .014).

A total of 722 patients were randomized to either ixazomib 4 mg or placebo weekly on days 1, 8, 15 and 22 of each 28-day cycle, with oral lenalidomide 25 mg on days 1 through 21 (with dose reductions in patients with renal impairment at the investigator’s discretion) and oral dexamethasone 40 mg on days 1, 8, 15, and 22.

Patients were stratified at randomization by the number of prior therapies (one vs. two or three), previous proteasome-inhibitor exposure, and by International Staging System for multiple myeloma stage I or II vs. III. Cycles were repeated until disease progression or until patients experienced unacceptable toxicity.

The overall median age of patients was 66 (range, 30-91) years. In all, 70% had received a prior proteasome inhibitor, 88% had International Staging System stage I or II disease, and 59% had received just one prior line of therapy.

Dr. Moreau noted that the responses were rapid and durable with a median time to response of 1.1 months for ixazomib and 1.9 months for placebo, and a median duration of response of 20.5 months and 15 months, respectively. Neither difference was significant.

The incidence of grade 3 or greater adverse events was 68% for patients on ixazomib vs. 61% with placebo. A higher incidence of thrombocytopenia with ixazomib (19% vs. 9%) primarily accounted for the difference.

Dr. Moreau noted that peripheral neuropathy, a common problem with intravenous proteasome inhibitors, was low with ixazomib, and patients did not report significant changes in their quality of life.

The study was sponsored by Millennium Pharmaceuticals. Dr. Moreau disclosed receiving honoraria from the company.

ORLANDO – The first all-oral drug combination for treatment of relapsed/refractory multiple myeloma significantly extended progression-free survival (PFS), based on the first of three planned interim analyses from a phase 3 trial.

The combination of the oral proteasome inhibitor ixazomib (Ninlaro, recently approved by the U.S. Food and Drug Administration) with lenalidomide and dexamethasone (len/dex) was associated with a 35% improvement in PFS, compared with len/dex and placebo, reported Dr. Philippe Moreau from Université Hospital of Nantes (France), Hôtel-Dieu.

The ixazomib combination also was associated with a significantly better median time to progression, at 21.4 vs. 15.7 months (P = .007). The time to response and duration of response were not significantly different, however, for the two groups in the placebo-controlled, randomized study of 722 patients.

“Ixazomib, when combined with len/dex for patients with relapsed and/or refractory multiple myeloma, was associated a significant and meaningful improvement in progression-free survival, significantly improved time to progression, and confirmed response rates,” Dr. Moreau said at a briefing at the American Society of Hematology annual meeting.

Dr. Moreau presented results of the phase III Tourmaline study, which compared len/dex plus weekly ixazomib or len/dex with weekly placebo in adults who have relapsed/refractory multiple myeloma following one to three prior lines of therapy and were not refractory to either prior lenalidomide or proteasome inhibitor–based therapy.

The study met its primary endpoint of a PFS advantage for the addition of ixazomib. The median PFS for ixazomib plus len/dex was 20.6 months, compared with 14.7 months for len/dex plus placebo. The hazard ratio for PFS with ixazomib-containing combination was 0.742 (P = .012).

The confirmed overall response rate (partial response or greater) was 78% with ixazomib vs. 71% with placebo (P = .035). The respective very good partial response or better rates were 48% vs. 39% (P = .014).

A total of 722 patients were randomized to either ixazomib 4 mg or placebo weekly on days 1, 8, 15 and 22 of each 28-day cycle, with oral lenalidomide 25 mg on days 1 through 21 (with dose reductions in patients with renal impairment at the investigator’s discretion) and oral dexamethasone 40 mg on days 1, 8, 15, and 22.

Patients were stratified at randomization by the number of prior therapies (one vs. two or three), previous proteasome-inhibitor exposure, and by International Staging System for multiple myeloma stage I or II vs. III. Cycles were repeated until disease progression or until patients experienced unacceptable toxicity.

The overall median age of patients was 66 (range, 30-91) years. In all, 70% had received a prior proteasome inhibitor, 88% had International Staging System stage I or II disease, and 59% had received just one prior line of therapy.

Dr. Moreau noted that the responses were rapid and durable with a median time to response of 1.1 months for ixazomib and 1.9 months for placebo, and a median duration of response of 20.5 months and 15 months, respectively. Neither difference was significant.

The incidence of grade 3 or greater adverse events was 68% for patients on ixazomib vs. 61% with placebo. A higher incidence of thrombocytopenia with ixazomib (19% vs. 9%) primarily accounted for the difference.

Dr. Moreau noted that peripheral neuropathy, a common problem with intravenous proteasome inhibitors, was low with ixazomib, and patients did not report significant changes in their quality of life.

The study was sponsored by Millennium Pharmaceuticals. Dr. Moreau disclosed receiving honoraria from the company.

References

References

Publications
Publications
Topics
Article Type
Display Headline
ASH: All-oral regimen extends multiple myeloma PFS
Display Headline
ASH: All-oral regimen extends multiple myeloma PFS
Sections
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

Vitals

<p><b>Key clinical point: </b>Ixazomib added to lenalidomide and dexamethasone is an effective all-oral drug regimen in relapsed/refractory multiple myeloma.
</p><p><b>Major finding:</b> The combination was associated with a 35% improvement in progression-free survival, compared with len/dex and placebo.<b></b>
</p><p><b>Data source: </b>Randomized placebo-controlled trial in 722 patients with relapsed/refractory multiple myeloma treated with one to three prior lines of therapy.
</p><p><b>Disclosures: </b>The study was sponsored by Millennium Pharmaceuticals. Dr. Moreau disclosed receiving honoraria from the company.</p>

VIDEO: Targeted agent, len/dex combo perform ‘impressively’ in advanced multiple myeloma

Article Type
Changed
Fri, 01/04/2019 - 09:46
Display Headline
VIDEO: Targeted agent, len/dex combo perform ‘impressively’ in advanced multiple myeloma

ORLANDO – A combination of daratumumab (Darzalex) with lenalidomide and dexamethasone (len/dex) was associated with a high overall response rate, including stringent complete responses, in patients with heavily pretreated relapsed/refractory multiple myeloma.

Daratumumab was recently approved by the Food and Drug Administration as monotherapy in patients with relapsed/refractory multiple myeloma.

Dr. Torben Plesner of Vejle Hospital and the University of Southern Denmark in Vejle, who dosed the first patient with daratumumab in 2007, discusses updated findings from a phase I/II trial of the daratumumab and len/dex combination.

tor@frontlinemedcom.com

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – A combination of daratumumab (Darzalex) with lenalidomide and dexamethasone (len/dex) was associated with a high overall response rate, including stringent complete responses, in patients with heavily pretreated relapsed/refractory multiple myeloma.

Daratumumab was recently approved by the Food and Drug Administration as monotherapy in patients with relapsed/refractory multiple myeloma.

Dr. Torben Plesner of Vejle Hospital and the University of Southern Denmark in Vejle, who dosed the first patient with daratumumab in 2007, discusses updated findings from a phase I/II trial of the daratumumab and len/dex combination.

tor@frontlinemedcom.com

ORLANDO – A combination of daratumumab (Darzalex) with lenalidomide and dexamethasone (len/dex) was associated with a high overall response rate, including stringent complete responses, in patients with heavily pretreated relapsed/refractory multiple myeloma.

Daratumumab was recently approved by the Food and Drug Administration as monotherapy in patients with relapsed/refractory multiple myeloma.

Dr. Torben Plesner of Vejle Hospital and the University of Southern Denmark in Vejle, who dosed the first patient with daratumumab in 2007, discusses updated findings from a phase I/II trial of the daratumumab and len/dex combination.

tor@frontlinemedcom.com

References

References

Publications
Publications
Topics
Article Type
Display Headline
VIDEO: Targeted agent, len/dex combo perform ‘impressively’ in advanced multiple myeloma
Display Headline
VIDEO: Targeted agent, len/dex combo perform ‘impressively’ in advanced multiple myeloma
Sections
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

ASH: Novel GBT440 reduces sickle cells, improves hematologic parameters

Article Type
Changed
Fri, 01/18/2019 - 15:30
Display Headline
ASH: Novel GBT440 reduces sickle cells, improves hematologic parameters

ORLANDO – An experimental agent that restores plasticity to red blood cells significantly improved hematologic parameters and reduced the deformation of red blood cells from sickle cell disease, suggest preliminary results from a phase I/II randomized clinical trial.

The drug, labeled GBT440, was well tolerated over one month, with no serious drug-related adverse events and no evidence of tissue hypoxia, said Dr. Claire Hemmaway from Queens Hospital in Essex, United Kingdom.

“We have emerging data with 28 days of dosing, and this supports the hypothesis that this drug inhibits polymerization of sickle hemoglobin, improves hemolysis, reduces red blood cell damage even in the micro-circulation, and improves oxygen delivery. Longer-term dosing is clearly required and this will define the optimal hematologic effects and the clinical benefit of this drug,” she said at the American Society of Hematology annual meeting.

RTEmagicC_f9456ca_Hemmanway_Claire_J_UK.jpg.jpg
Dr. Claire Hemenway

Although its mechanism of action is not completely understood, GBT440 is a small-molecule hemoglobin modifier which is known to increase hemoglobin oxygen affinity. This first-in-class agent has been shown in both in vitro and in vivo studies to be a strong and direct anti-sickling agent. The drug has also been shown to inhibit polymerization of hemoglobin S, the central event in sickle cell disease, Dr. Hemmaway said.

“We hypothesized that patients on GBT440 will see a reduction in the red cell damage, see a reduction in hemolysis, and an improvement in the anemia, and with an inhibition of the formation of the sickle cells we’ll also see an improvement in blood flow, and this could potentially modify the course of sickle cell disease in our patients,” she said at a briefing prior to her presentation of the data in an oral abstract session.

To test the safety and efficacy of the drug, the investigators enrolled 64 healthy volunteers and 16 patients with homozygous HbSS sickle-cell disease into a phase I/II randomized, double-placebo controlled, parallel group trial.  

The patients all had baseline hemoglobin levels from 6 to 10 g/dL, and had not had a vaso-occlusive crisis or transfusion with 30 days of screening.

The study was divided into parts, with part A testing single ascending doses, and part B testing multiple ascending doses compared with placebo on a 6:2 randomization basis.

As of July 24 2015, 54 healthy volunteers had completed the study, two were discontinued due to mild-to-moderate rash and/or headache, and eight were still on follow-up.

Eight patients with sickle cell disease completed part A, and eight were on follow-up in part B. No patients dropped out of the study, although one had a dose reduction from 700 mg to 400 mg because of abdominal discomfort.

Patients and volunteers generally tolerated the drug well, with generally mild adverse events, no deaths, and just one serious adverse event, an acute painful crisis in a patient on placebo.

The drug was shown to increase hemoglobin in both healthy volunteers and patients, reduce reticulocytosis, and improve biomarkers of hemolysis and inflammation. The hematologic effects correlate with GBT440 blood levels, Dr. Hemmaway said.

“You can see dramatic reductions in the reticulocyte counts, which maximally are reduced by more than 50%, and these are maintained over 28 days. The reduction in reticulocyte counts suggests an improvement in red cell life span,” she said.

One patient who received a 700 mg daily oral dose of GBT440 had evidence of a complete absence of sickled cells 28 days after starting on therapy, she noted.

The investigators have not been able to determine whether the hematologic improvements seen in the study correlate with symptomatic improvements, because they have only 28 days of dosing data and the results are still blinded, Dr. Hemmaway said.

A pediatric hematologist who was not involved in the study said that the early results offer hope for patients.

“I think many of us in the field continue to come back to the painful reality that as of today there is only one FDA-approved medication for sickle cell disease. That fundamentally is something that I think many of us are extremely troubled by, especially at a meeting like this where we see so many other opportunities in other conditions,” commented Dr. Alexis Thompson, a professor of hematology at the Robert H. Lurie Children’s Hospital of Chicago, Illinois.

Although the data are early, they are encouraging enough to justify moving forward with a phase II trial, which is planned to start in early 2016.

“I think the person who is going to be the ultimate beneficiary from many of these advances will be a child who can look forward to not only a longer lifespan but a lifespan that is less hampered by the complications of this disease,” she said.

 

 

Dr. Thompson moderated the briefing in which Dr. Hemmaway presented the data.

The study is supported by Global Blood Therapeutics. Dr. Hemmaway had no relevant disclosures. Several co-investigators are employees of the company. Dr, Thompson had no disclosures relevant to the study.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – An experimental agent that restores plasticity to red blood cells significantly improved hematologic parameters and reduced the deformation of red blood cells from sickle cell disease, suggest preliminary results from a phase I/II randomized clinical trial.

The drug, labeled GBT440, was well tolerated over one month, with no serious drug-related adverse events and no evidence of tissue hypoxia, said Dr. Claire Hemmaway from Queens Hospital in Essex, United Kingdom.

“We have emerging data with 28 days of dosing, and this supports the hypothesis that this drug inhibits polymerization of sickle hemoglobin, improves hemolysis, reduces red blood cell damage even in the micro-circulation, and improves oxygen delivery. Longer-term dosing is clearly required and this will define the optimal hematologic effects and the clinical benefit of this drug,” she said at the American Society of Hematology annual meeting.

RTEmagicC_f9456ca_Hemmanway_Claire_J_UK.jpg.jpg
Dr. Claire Hemenway

Although its mechanism of action is not completely understood, GBT440 is a small-molecule hemoglobin modifier which is known to increase hemoglobin oxygen affinity. This first-in-class agent has been shown in both in vitro and in vivo studies to be a strong and direct anti-sickling agent. The drug has also been shown to inhibit polymerization of hemoglobin S, the central event in sickle cell disease, Dr. Hemmaway said.

“We hypothesized that patients on GBT440 will see a reduction in the red cell damage, see a reduction in hemolysis, and an improvement in the anemia, and with an inhibition of the formation of the sickle cells we’ll also see an improvement in blood flow, and this could potentially modify the course of sickle cell disease in our patients,” she said at a briefing prior to her presentation of the data in an oral abstract session.

To test the safety and efficacy of the drug, the investigators enrolled 64 healthy volunteers and 16 patients with homozygous HbSS sickle-cell disease into a phase I/II randomized, double-placebo controlled, parallel group trial.  

The patients all had baseline hemoglobin levels from 6 to 10 g/dL, and had not had a vaso-occlusive crisis or transfusion with 30 days of screening.

The study was divided into parts, with part A testing single ascending doses, and part B testing multiple ascending doses compared with placebo on a 6:2 randomization basis.

As of July 24 2015, 54 healthy volunteers had completed the study, two were discontinued due to mild-to-moderate rash and/or headache, and eight were still on follow-up.

Eight patients with sickle cell disease completed part A, and eight were on follow-up in part B. No patients dropped out of the study, although one had a dose reduction from 700 mg to 400 mg because of abdominal discomfort.

Patients and volunteers generally tolerated the drug well, with generally mild adverse events, no deaths, and just one serious adverse event, an acute painful crisis in a patient on placebo.

The drug was shown to increase hemoglobin in both healthy volunteers and patients, reduce reticulocytosis, and improve biomarkers of hemolysis and inflammation. The hematologic effects correlate with GBT440 blood levels, Dr. Hemmaway said.

“You can see dramatic reductions in the reticulocyte counts, which maximally are reduced by more than 50%, and these are maintained over 28 days. The reduction in reticulocyte counts suggests an improvement in red cell life span,” she said.

One patient who received a 700 mg daily oral dose of GBT440 had evidence of a complete absence of sickled cells 28 days after starting on therapy, she noted.

The investigators have not been able to determine whether the hematologic improvements seen in the study correlate with symptomatic improvements, because they have only 28 days of dosing data and the results are still blinded, Dr. Hemmaway said.

A pediatric hematologist who was not involved in the study said that the early results offer hope for patients.

“I think many of us in the field continue to come back to the painful reality that as of today there is only one FDA-approved medication for sickle cell disease. That fundamentally is something that I think many of us are extremely troubled by, especially at a meeting like this where we see so many other opportunities in other conditions,” commented Dr. Alexis Thompson, a professor of hematology at the Robert H. Lurie Children’s Hospital of Chicago, Illinois.

Although the data are early, they are encouraging enough to justify moving forward with a phase II trial, which is planned to start in early 2016.

“I think the person who is going to be the ultimate beneficiary from many of these advances will be a child who can look forward to not only a longer lifespan but a lifespan that is less hampered by the complications of this disease,” she said.

 

 

Dr. Thompson moderated the briefing in which Dr. Hemmaway presented the data.

The study is supported by Global Blood Therapeutics. Dr. Hemmaway had no relevant disclosures. Several co-investigators are employees of the company. Dr, Thompson had no disclosures relevant to the study.

ORLANDO – An experimental agent that restores plasticity to red blood cells significantly improved hematologic parameters and reduced the deformation of red blood cells from sickle cell disease, suggest preliminary results from a phase I/II randomized clinical trial.

The drug, labeled GBT440, was well tolerated over one month, with no serious drug-related adverse events and no evidence of tissue hypoxia, said Dr. Claire Hemmaway from Queens Hospital in Essex, United Kingdom.

“We have emerging data with 28 days of dosing, and this supports the hypothesis that this drug inhibits polymerization of sickle hemoglobin, improves hemolysis, reduces red blood cell damage even in the micro-circulation, and improves oxygen delivery. Longer-term dosing is clearly required and this will define the optimal hematologic effects and the clinical benefit of this drug,” she said at the American Society of Hematology annual meeting.

RTEmagicC_f9456ca_Hemmanway_Claire_J_UK.jpg.jpg
Dr. Claire Hemenway

Although its mechanism of action is not completely understood, GBT440 is a small-molecule hemoglobin modifier which is known to increase hemoglobin oxygen affinity. This first-in-class agent has been shown in both in vitro and in vivo studies to be a strong and direct anti-sickling agent. The drug has also been shown to inhibit polymerization of hemoglobin S, the central event in sickle cell disease, Dr. Hemmaway said.

“We hypothesized that patients on GBT440 will see a reduction in the red cell damage, see a reduction in hemolysis, and an improvement in the anemia, and with an inhibition of the formation of the sickle cells we’ll also see an improvement in blood flow, and this could potentially modify the course of sickle cell disease in our patients,” she said at a briefing prior to her presentation of the data in an oral abstract session.

To test the safety and efficacy of the drug, the investigators enrolled 64 healthy volunteers and 16 patients with homozygous HbSS sickle-cell disease into a phase I/II randomized, double-placebo controlled, parallel group trial.  

The patients all had baseline hemoglobin levels from 6 to 10 g/dL, and had not had a vaso-occlusive crisis or transfusion with 30 days of screening.

The study was divided into parts, with part A testing single ascending doses, and part B testing multiple ascending doses compared with placebo on a 6:2 randomization basis.

As of July 24 2015, 54 healthy volunteers had completed the study, two were discontinued due to mild-to-moderate rash and/or headache, and eight were still on follow-up.

Eight patients with sickle cell disease completed part A, and eight were on follow-up in part B. No patients dropped out of the study, although one had a dose reduction from 700 mg to 400 mg because of abdominal discomfort.

Patients and volunteers generally tolerated the drug well, with generally mild adverse events, no deaths, and just one serious adverse event, an acute painful crisis in a patient on placebo.

The drug was shown to increase hemoglobin in both healthy volunteers and patients, reduce reticulocytosis, and improve biomarkers of hemolysis and inflammation. The hematologic effects correlate with GBT440 blood levels, Dr. Hemmaway said.

“You can see dramatic reductions in the reticulocyte counts, which maximally are reduced by more than 50%, and these are maintained over 28 days. The reduction in reticulocyte counts suggests an improvement in red cell life span,” she said.

One patient who received a 700 mg daily oral dose of GBT440 had evidence of a complete absence of sickled cells 28 days after starting on therapy, she noted.

The investigators have not been able to determine whether the hematologic improvements seen in the study correlate with symptomatic improvements, because they have only 28 days of dosing data and the results are still blinded, Dr. Hemmaway said.

A pediatric hematologist who was not involved in the study said that the early results offer hope for patients.

“I think many of us in the field continue to come back to the painful reality that as of today there is only one FDA-approved medication for sickle cell disease. That fundamentally is something that I think many of us are extremely troubled by, especially at a meeting like this where we see so many other opportunities in other conditions,” commented Dr. Alexis Thompson, a professor of hematology at the Robert H. Lurie Children’s Hospital of Chicago, Illinois.

Although the data are early, they are encouraging enough to justify moving forward with a phase II trial, which is planned to start in early 2016.

“I think the person who is going to be the ultimate beneficiary from many of these advances will be a child who can look forward to not only a longer lifespan but a lifespan that is less hampered by the complications of this disease,” she said.

 

 

Dr. Thompson moderated the briefing in which Dr. Hemmaway presented the data.

The study is supported by Global Blood Therapeutics. Dr. Hemmaway had no relevant disclosures. Several co-investigators are employees of the company. Dr, Thompson had no disclosures relevant to the study.

References

References

Publications
Publications
Topics
Article Type
Display Headline
ASH: Novel GBT440 reduces sickle cells, improves hematologic parameters
Display Headline
ASH: Novel GBT440 reduces sickle cells, improves hematologic parameters
Sections
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

Vitals

<p><b>Key clinical point:</b> A first-in class small molecule drug improved hematologic parameters and reduced sickling of red blood cells.
</p><p><b>Major finding:</b> GBT440 inhibits polymerization of sickle hemoglobin, improves hemolysis, reduces red blood cell damage, and improves oxygen delivery.
</p><p><b>Data source:</b> Randomized, double blind, placebo-controlled phase I/II trial.
</p><p><b>Disclosures:</b> The study is supported by Global Blood Therapeutics. Dr. Hemmaway had no relevant disclosures. Several co-investigators are employees of the company. Dr, Thompson had no disclosures relevant to the study.</p>

ASH: HLA-identical sibling transplants “excellent” in eligble SCD patients

Article Type
Changed
Fri, 01/18/2019 - 15:30
Display Headline
ASH: HLA-identical sibling transplants “excellent” in eligble SCD patients

ORLANDO – For patients with severe sickle cell disease, stem cell transplants from an HLA identical sibling are associated with “excellent” 3-year overall and event-free survival, results of a retrospective analysis show.

Data on 1,000 hematopoietic stem cell transplants (HSCT) in children and adults with sickle-cell disease performed in 88 centers in 23 countries showed a 3-year event-free-survival (EFS) rate of 90%, and 3-year overall survival (OS) rate of 94%, reported Dr. Barbara Cappelli from the Eurocord International Registry in Paris, France.

RTEmagicC_a7ddd27_Cappelli_Barbara_France.jpg.jpg
Dr. Barbara Cappelli

“Early referral to transplant for patients with severe sickle-cell disease is warranted, as age is an independent predictor for both event-free survival and overall survival,” she said about the study at a briefing at the American Society of Hematology annual meeting.

Patients who received donor cord blood had the best 3-year OS, at 99%, compared with 94% for patients who received bone marrow grafts, and 80% for those who received peripheral blood stem cells (PBSC).

“Transplants from peripheral blood are not recommended, because the result that there is higher mortality with transplants with peripheral blood,” she said.

The findings provide further evidence that for patients with severe sickle-disease who have an HLA-identical sibling available as a donor, HSCT can be curative with good safety, commented Dr. Alexis Thompson, a professor of hematology at the Robert H. Lurie Children’s Hospital of Chicago, Illinois.

Dr. Thompson was not involved in the study, but moderated a briefing where the data were presented.

International Registries

HSCT is the only therapy currently known to be curative for sickle-cell disease, but due to the risk for early and late complications it is generally reserved for patients with the most severe anemia or most disabling symptoms.

To evaluate the efficacy and safety of HSCT for sickle-cell disease, the investigators took a retrospective look at data from bone marrow transplant registries covering Europe, Brazil, the United States, and other reporting centers.

They searched for data on all patients with sickle-cell disease, children and adults, who received a transplant from an HLA identical sibling from 1986 through 2013.

The majority of patients (85%) were younger than 16, with the median age at HSCT of 9 years (range 1 to 54 years). Stroke was the most common indication for HSCT.

In all, 87% of patients received a myeloablative conditioning regimen based on busulfan combined with either cyclophosphamide or fludarabine. The remaining 13% of patients underwent a reduced-intensity conditioning regimen, primarily with fludarabine and cyclophosphamide.

In vivo T-cell depletion was performed in 70% of patients, either with anti-thymocyte globulin (630 patients) or with alemtumumab (76).

Prophylaxis for graft vs. host disease (GVHD) was predominantly cyclosporine as monotherapy or in combination with methotrexate.

A large majority of patients (84%) received bone marrow grafts, 7% got PBSCs, and 9% received umbilical cord blood.

By 60 days, the cumulative incidence of engraftment was 98%, with a median cumulative incidence of platelet engraftment of 25 days.

The cumulative incidence of acute GVHD (within 100 days of transplant) 14.3%, and the cumulative incidence of chronic GVHD (out to 3 years) was 13.3%

In all, 71 patients (7%) required autologous reconstitution, 45 because of late graft failure, 31 patients (3%) underwent a second transplant and 67 patients (7%) died. Of the patients who died, 6% had received bone marrow, 1% had received cord blood, and 21% had received PBSC.

As noted before, respective EFS and OS 3 years after transplant were 90% and 94%.

In multivariate analysis controlled for transplant and demographic characteristics, the authors found that younger age at transplant and use of bone marrow or cord blood were independently associated with better EFS and OS. In addition, 3-year overall survival was significantly better among patients who received transplants from the year 2000 on.

The risk for acute GVHD was significantly associated with increasing age, but none of the variables tested were associated with chronic GVHD.

Dr. Cappelli said that ways to improve outcomes include performing pre-or post-natal diagnosis in at-risk families, performing HLA typing of all family members at the time of diagnosis, and going to transplant as soon as the criteria for severe sickle-cell disease are met.

The study was supported by Eurocord-Monacord, EBMT Paediatric Disease Working Party and CIBMTR. Dr. Cappelli and Dr. Johnson reported having no conflicts of interest.

References

Meeting/Event
Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

ORLANDO – For patients with severe sickle cell disease, stem cell transplants from an HLA identical sibling are associated with “excellent” 3-year overall and event-free survival, results of a retrospective analysis show.

Data on 1,000 hematopoietic stem cell transplants (HSCT) in children and adults with sickle-cell disease performed in 88 centers in 23 countries showed a 3-year event-free-survival (EFS) rate of 90%, and 3-year overall survival (OS) rate of 94%, reported Dr. Barbara Cappelli from the Eurocord International Registry in Paris, France.

RTEmagicC_a7ddd27_Cappelli_Barbara_France.jpg.jpg
Dr. Barbara Cappelli

“Early referral to transplant for patients with severe sickle-cell disease is warranted, as age is an independent predictor for both event-free survival and overall survival,” she said about the study at a briefing at the American Society of Hematology annual meeting.

Patients who received donor cord blood had the best 3-year OS, at 99%, compared with 94% for patients who received bone marrow grafts, and 80% for those who received peripheral blood stem cells (PBSC).

“Transplants from peripheral blood are not recommended, because the result that there is higher mortality with transplants with peripheral blood,” she said.

The findings provide further evidence that for patients with severe sickle-disease who have an HLA-identical sibling available as a donor, HSCT can be curative with good safety, commented Dr. Alexis Thompson, a professor of hematology at the Robert H. Lurie Children’s Hospital of Chicago, Illinois.

Dr. Thompson was not involved in the study, but moderated a briefing where the data were presented.

International Registries

HSCT is the only therapy currently known to be curative for sickle-cell disease, but due to the risk for early and late complications it is generally reserved for patients with the most severe anemia or most disabling symptoms.

To evaluate the efficacy and safety of HSCT for sickle-cell disease, the investigators took a retrospective look at data from bone marrow transplant registries covering Europe, Brazil, the United States, and other reporting centers.

They searched for data on all patients with sickle-cell disease, children and adults, who received a transplant from an HLA identical sibling from 1986 through 2013.

The majority of patients (85%) were younger than 16, with the median age at HSCT of 9 years (range 1 to 54 years). Stroke was the most common indication for HSCT.

In all, 87% of patients received a myeloablative conditioning regimen based on busulfan combined with either cyclophosphamide or fludarabine. The remaining 13% of patients underwent a reduced-intensity conditioning regimen, primarily with fludarabine and cyclophosphamide.

In vivo T-cell depletion was performed in 70% of patients, either with anti-thymocyte globulin (630 patients) or with alemtumumab (76).

Prophylaxis for graft vs. host disease (GVHD) was predominantly cyclosporine as monotherapy or in combination with methotrexate.

A large majority of patients (84%) received bone marrow grafts, 7% got PBSCs, and 9% received umbilical cord blood.

By 60 days, the cumulative incidence of engraftment was 98%, with a median cumulative incidence of platelet engraftment of 25 days.

The cumulative incidence of acute GVHD (within 100 days of transplant) 14.3%, and the cumulative incidence of chronic GVHD (out to 3 years) was 13.3%

In all, 71 patients (7%) required autologous reconstitution, 45 because of late graft failure, 31 patients (3%) underwent a second transplant and 67 patients (7%) died. Of the patients who died, 6% had received bone marrow, 1% had received cord blood, and 21% had received PBSC.

As noted before, respective EFS and OS 3 years after transplant were 90% and 94%.

In multivariate analysis controlled for transplant and demographic characteristics, the authors found that younger age at transplant and use of bone marrow or cord blood were independently associated with better EFS and OS. In addition, 3-year overall survival was significantly better among patients who received transplants from the year 2000 on.

The risk for acute GVHD was significantly associated with increasing age, but none of the variables tested were associated with chronic GVHD.

Dr. Cappelli said that ways to improve outcomes include performing pre-or post-natal diagnosis in at-risk families, performing HLA typing of all family members at the time of diagnosis, and going to transplant as soon as the criteria for severe sickle-cell disease are met.

The study was supported by Eurocord-Monacord, EBMT Paediatric Disease Working Party and CIBMTR. Dr. Cappelli and Dr. Johnson reported having no conflicts of interest.

ORLANDO – For patients with severe sickle cell disease, stem cell transplants from an HLA identical sibling are associated with “excellent” 3-year overall and event-free survival, results of a retrospective analysis show.

Data on 1,000 hematopoietic stem cell transplants (HSCT) in children and adults with sickle-cell disease performed in 88 centers in 23 countries showed a 3-year event-free-survival (EFS) rate of 90%, and 3-year overall survival (OS) rate of 94%, reported Dr. Barbara Cappelli from the Eurocord International Registry in Paris, France.

RTEmagicC_a7ddd27_Cappelli_Barbara_France.jpg.jpg
Dr. Barbara Cappelli

“Early referral to transplant for patients with severe sickle-cell disease is warranted, as age is an independent predictor for both event-free survival and overall survival,” she said about the study at a briefing at the American Society of Hematology annual meeting.

Patients who received donor cord blood had the best 3-year OS, at 99%, compared with 94% for patients who received bone marrow grafts, and 80% for those who received peripheral blood stem cells (PBSC).

“Transplants from peripheral blood are not recommended, because the result that there is higher mortality with transplants with peripheral blood,” she said.

The findings provide further evidence that for patients with severe sickle-disease who have an HLA-identical sibling available as a donor, HSCT can be curative with good safety, commented Dr. Alexis Thompson, a professor of hematology at the Robert H. Lurie Children’s Hospital of Chicago, Illinois.

Dr. Thompson was not involved in the study, but moderated a briefing where the data were presented.

International Registries

HSCT is the only therapy currently known to be curative for sickle-cell disease, but due to the risk for early and late complications it is generally reserved for patients with the most severe anemia or most disabling symptoms.

To evaluate the efficacy and safety of HSCT for sickle-cell disease, the investigators took a retrospective look at data from bone marrow transplant registries covering Europe, Brazil, the United States, and other reporting centers.

They searched for data on all patients with sickle-cell disease, children and adults, who received a transplant from an HLA identical sibling from 1986 through 2013.

The majority of patients (85%) were younger than 16, with the median age at HSCT of 9 years (range 1 to 54 years). Stroke was the most common indication for HSCT.

In all, 87% of patients received a myeloablative conditioning regimen based on busulfan combined with either cyclophosphamide or fludarabine. The remaining 13% of patients underwent a reduced-intensity conditioning regimen, primarily with fludarabine and cyclophosphamide.

In vivo T-cell depletion was performed in 70% of patients, either with anti-thymocyte globulin (630 patients) or with alemtumumab (76).

Prophylaxis for graft vs. host disease (GVHD) was predominantly cyclosporine as monotherapy or in combination with methotrexate.

A large majority of patients (84%) received bone marrow grafts, 7% got PBSCs, and 9% received umbilical cord blood.

By 60 days, the cumulative incidence of engraftment was 98%, with a median cumulative incidence of platelet engraftment of 25 days.

The cumulative incidence of acute GVHD (within 100 days of transplant) 14.3%, and the cumulative incidence of chronic GVHD (out to 3 years) was 13.3%

In all, 71 patients (7%) required autologous reconstitution, 45 because of late graft failure, 31 patients (3%) underwent a second transplant and 67 patients (7%) died. Of the patients who died, 6% had received bone marrow, 1% had received cord blood, and 21% had received PBSC.

As noted before, respective EFS and OS 3 years after transplant were 90% and 94%.

In multivariate analysis controlled for transplant and demographic characteristics, the authors found that younger age at transplant and use of bone marrow or cord blood were independently associated with better EFS and OS. In addition, 3-year overall survival was significantly better among patients who received transplants from the year 2000 on.

The risk for acute GVHD was significantly associated with increasing age, but none of the variables tested were associated with chronic GVHD.

Dr. Cappelli said that ways to improve outcomes include performing pre-or post-natal diagnosis in at-risk families, performing HLA typing of all family members at the time of diagnosis, and going to transplant as soon as the criteria for severe sickle-cell disease are met.

The study was supported by Eurocord-Monacord, EBMT Paediatric Disease Working Party and CIBMTR. Dr. Cappelli and Dr. Johnson reported having no conflicts of interest.

References

References

Publications
Publications
Topics
Article Type
Display Headline
ASH: HLA-identical sibling transplants “excellent” in eligble SCD patients
Display Headline
ASH: HLA-identical sibling transplants “excellent” in eligble SCD patients
Sections
Article Source

AT ASH 2015

PURLs Copyright

Inside the Article

Vitals

<p><b>Key clinical point:</b><b> </b>Bone marrow transplants from HLA-identical siblings are frequently curative of sickle-cell disease.
</p><p><b>Major finding:</b><b> </b>Three-year event-free survival among 1000 patients was 90%, and 3-year overall survival was 94%.
</p><p><b> Data source:</b><b> </b>Retrospective review of registry data on 1000 patients who underwent hematopoietic stem cell transplants with HLA-identical sibling donors.
</p><p><b>Disclosures:</b><b> </b>The study was supported by<b> </b>Eurocord-Monacord, EBMT Paediatric Disease Working Party and CIBMTR. Dr. Cappelli and Dr. Johnson reported having no conflicts of interest.</p>