User login
FDA approves Nivestym, second biosimilar to Neupogen
Nivestym (filgrastim-aafi), a biosimilar to Neupogen (filgrastim) was approved July 20 by the Food and Drug Administration, according to a statement provided by the agency. Nivestym is the second biosimilar to Neupogen to be approved in the United States.
- Patients with cancer receiving myelosuppressive chemotherapy.
- Patients with acute myeloid leukemia receiving induction or consolidation chemotherapy.
- Patients with cancer undergoing bone marrow transplantation.
- Patients undergoing autologous peripheral blood progenitor cell collection and therapy.
- Patients with severe chronic neutropenia.
According to a press release from Pfizer, the manufacturer of the biosimilar, Nivestym is expected to be available in the United States at a significant discount to the current wholesale acquisition cost of Neupogen, which is not inclusive of discounts to payers, providers, distributors, and other purchasing organizations.
The FDA statement notes that a biosimilar is approved based on a showing that it is highly similar to an already approved biologic product, known as a reference product. The biosimilar also must be shown to have no clinically meaningful differences in terms of safety and effectiveness from the reference product. Only minor differences in clinically inactive components are allowable in biosimilar products.
Prescribing information is available here.
Nivestym (filgrastim-aafi), a biosimilar to Neupogen (filgrastim) was approved July 20 by the Food and Drug Administration, according to a statement provided by the agency. Nivestym is the second biosimilar to Neupogen to be approved in the United States.
- Patients with cancer receiving myelosuppressive chemotherapy.
- Patients with acute myeloid leukemia receiving induction or consolidation chemotherapy.
- Patients with cancer undergoing bone marrow transplantation.
- Patients undergoing autologous peripheral blood progenitor cell collection and therapy.
- Patients with severe chronic neutropenia.
According to a press release from Pfizer, the manufacturer of the biosimilar, Nivestym is expected to be available in the United States at a significant discount to the current wholesale acquisition cost of Neupogen, which is not inclusive of discounts to payers, providers, distributors, and other purchasing organizations.
The FDA statement notes that a biosimilar is approved based on a showing that it is highly similar to an already approved biologic product, known as a reference product. The biosimilar also must be shown to have no clinically meaningful differences in terms of safety and effectiveness from the reference product. Only minor differences in clinically inactive components are allowable in biosimilar products.
Prescribing information is available here.
Nivestym (filgrastim-aafi), a biosimilar to Neupogen (filgrastim) was approved July 20 by the Food and Drug Administration, according to a statement provided by the agency. Nivestym is the second biosimilar to Neupogen to be approved in the United States.
- Patients with cancer receiving myelosuppressive chemotherapy.
- Patients with acute myeloid leukemia receiving induction or consolidation chemotherapy.
- Patients with cancer undergoing bone marrow transplantation.
- Patients undergoing autologous peripheral blood progenitor cell collection and therapy.
- Patients with severe chronic neutropenia.
According to a press release from Pfizer, the manufacturer of the biosimilar, Nivestym is expected to be available in the United States at a significant discount to the current wholesale acquisition cost of Neupogen, which is not inclusive of discounts to payers, providers, distributors, and other purchasing organizations.
The FDA statement notes that a biosimilar is approved based on a showing that it is highly similar to an already approved biologic product, known as a reference product. The biosimilar also must be shown to have no clinically meaningful differences in terms of safety and effectiveness from the reference product. Only minor differences in clinically inactive components are allowable in biosimilar products.
Prescribing information is available here.
FDA approves IDH1 inhibitor for relapsed/refractory AML
The Food and Drug Administration has approved ivosidenib (Tibsovo) as the first treatment of adult patients with relapsed/refractory acute myeloid leukemia (AML) and an isocitrate dehydrogenase-1 (IDH1) mutation.
More specifically, the oral treatment has been approved for patients whose mutations have been identified by the Abbott RealTime IDH1 assay, a companion diagnostic test.
The approval was based on results from a phase 1, open-label, single-arm, multicenter, dose-escalation and expansion trial of adult patients in this AML population. The primary end point was combined complete remission and complete remission with partial hematologic improvement; this combined rate was 32.8%, and the median duration of this remission was 8.2 months.
The most serious adverse events included differentiation syndrome, QTc prolongation, and Guillain-Barré syndrome. Other adverse reactions included fatigue, leukocytosis, arthralgia, diarrhea, dyspnea, edema, and constipation.
Ivosidenib is marketed as Tibsovo by Agios Pharmaceuticals. The RealTime IDH1 Assay is marketed by Abbott Laboratories.
The Food and Drug Administration has approved ivosidenib (Tibsovo) as the first treatment of adult patients with relapsed/refractory acute myeloid leukemia (AML) and an isocitrate dehydrogenase-1 (IDH1) mutation.
More specifically, the oral treatment has been approved for patients whose mutations have been identified by the Abbott RealTime IDH1 assay, a companion diagnostic test.
The approval was based on results from a phase 1, open-label, single-arm, multicenter, dose-escalation and expansion trial of adult patients in this AML population. The primary end point was combined complete remission and complete remission with partial hematologic improvement; this combined rate was 32.8%, and the median duration of this remission was 8.2 months.
The most serious adverse events included differentiation syndrome, QTc prolongation, and Guillain-Barré syndrome. Other adverse reactions included fatigue, leukocytosis, arthralgia, diarrhea, dyspnea, edema, and constipation.
Ivosidenib is marketed as Tibsovo by Agios Pharmaceuticals. The RealTime IDH1 Assay is marketed by Abbott Laboratories.
The Food and Drug Administration has approved ivosidenib (Tibsovo) as the first treatment of adult patients with relapsed/refractory acute myeloid leukemia (AML) and an isocitrate dehydrogenase-1 (IDH1) mutation.
More specifically, the oral treatment has been approved for patients whose mutations have been identified by the Abbott RealTime IDH1 assay, a companion diagnostic test.
The approval was based on results from a phase 1, open-label, single-arm, multicenter, dose-escalation and expansion trial of adult patients in this AML population. The primary end point was combined complete remission and complete remission with partial hematologic improvement; this combined rate was 32.8%, and the median duration of this remission was 8.2 months.
The most serious adverse events included differentiation syndrome, QTc prolongation, and Guillain-Barré syndrome. Other adverse reactions included fatigue, leukocytosis, arthralgia, diarrhea, dyspnea, edema, and constipation.
Ivosidenib is marketed as Tibsovo by Agios Pharmaceuticals. The RealTime IDH1 Assay is marketed by Abbott Laboratories.
Diabetics have higher risk of hematologic, other cancers
A review of data from more than 19 million people indicates that diabetes significantly raises a person’s risk of developing cancer.
When researchers compared patients with diabetes and without, both male and female diabetics had an increased risk of leukemias and lymphomas as well as certain solid tumors.
Researchers also found that diabetes conferred a higher cancer risk for women than men, both for all cancers combined and for some specific cancers, including leukemia.
“The link between diabetes and the risk of developing cancer is now firmly established,” said Toshiaki Ohkuma, PhD, of The George Institute for Global Health at the University of New South Wales in Australia.
“We have also demonstrated, for the first time, that women with diabetes are more likely to develop any form of cancer and have a significantly higher chance of developing kidney, oral, and stomach cancers and leukemia.”
Dr Ohkuma and his colleagues reported these findings in Diabetologia.
The researchers conducted a systematic search in PubMed MEDLINE to identify reports on the links between diabetes and cancer. Additional reports were identified from the reference lists of the relevant studies.
Only those cohort studies providing relative risks (RRs) for the association between diabetes and cancer for both women and men were included. In total, 107 relevant articles were identified, along with 36 cohorts of individual participant data.
RRs for cancer were obtained for patients with diabetes (types 1 and 2 combined) versus those without diabetes, for both men and women. The women-to-men ratios of these relative risk ratios (RRRs) were then calculated to determine the excess risk in women if present.
Data on all-site cancer was available from 47 studies, involving 121 cohorts and 19,239,302 individuals.
Diabetics vs non-diabetics
Women with diabetes had a 27% higher risk of all-site cancer compared to women without diabetes (RR=1.27; 95% CI 1.21, 1.32; P<0.001).
For men, the risk of all-site cancer was 19% higher among those with diabetes than those without (RR=1.19; 95% CI 1.13, 1.25; P<0.001).
There were several hematologic malignancies for which diabetics had an increased risk, as shown in the following table.
Cancer type | RR for women (99% CI) | RR for men (99% CI) |
Lymphatic and hematopoietic tissue | 1.24 (1.05, 1.46)* | 1.21 (0.98, 1.48) |
Leukemia | 1.53 (1.00, 2.33) | 1.22 (0.80, 1.85) |
Myeloid leukemia | 0.83 (0.39, 1.76) | 1.12 (0.77, 1.62) |
Acute myeloid leukemia | 1.33 (1.12, 1.57)* | 1.14 (0.56, 2.33) |
Chronic myeloid leukemia | 1.67 (1.27, 2.20)* | 1.62 (1.32, 1.98)* |
Lymphoid leukemia | 1.74 (0.31, 9.79) | 1.20 (0.86, 1.68) |
Lymphoma | 2.31 (0.57, 9.30) | 1.80 (0.68, 4.75) |
Non-Hodgkin lymphoma | 1.16 (1.02, 1.32)* | 1.20 (1.08, 1.34)* |
Hodgkin lymphoma | 1.20 (0.61, 2.38) | 1.36 (1.05, 1.77)* |
Multiple myeloma | 1.19 (0.97, 1.47) | 1.12 (0.90, 1.41) |
*denotes statistical significance with a P value < 0.01 |
Sex comparison
Calculation of the women-to-men ratio revealed that women with diabetes had a 6% greater excess risk of all-site cancer compared to men with diabetes (RRR=1.06; 95% CI 1.03, 1.09; P<0.001).
The women-to-men ratios also showed significantly higher risks for female diabetics for:
- Kidney cancer—RRR=1.11 (99% CI 1.04, 1.18; P<0.001)
- Oral cancer—RRR=1.13 (99% CI 1.00, 1.28; P=0.009)
- Stomach cancer—RRR=1.14 (99% CI 1.07, 1.22; P<0.001)
- Leukemia—RRR=1.15 (99% CI 1.02, 1.28; P=0.002).
However, women had a significantly lower risk of liver cancer (RRR=0.88; 99% CI 0.79, 0.99; P=0.005).
There are several possible reasons for the excess cancer risk observed in women, according to study author Sanne Peters, PhD, of The George Institute for Global Health at the University of Oxford in the UK.
For example, on average, women are in the pre-diabetic state of impaired glucose tolerance 2 years longer than men.
“Historically, we know that women are often under-treated when they first present with symptoms of diabetes, are less likely to receive intensive care, and are not taking the same levels of medications as men,” Dr Peters said. “All of these could go some way into explaining why women are at greater risk of developing cancer, but, without more research, we can’t be certain.”
A review of data from more than 19 million people indicates that diabetes significantly raises a person’s risk of developing cancer.
When researchers compared patients with diabetes and without, both male and female diabetics had an increased risk of leukemias and lymphomas as well as certain solid tumors.
Researchers also found that diabetes conferred a higher cancer risk for women than men, both for all cancers combined and for some specific cancers, including leukemia.
“The link between diabetes and the risk of developing cancer is now firmly established,” said Toshiaki Ohkuma, PhD, of The George Institute for Global Health at the University of New South Wales in Australia.
“We have also demonstrated, for the first time, that women with diabetes are more likely to develop any form of cancer and have a significantly higher chance of developing kidney, oral, and stomach cancers and leukemia.”
Dr Ohkuma and his colleagues reported these findings in Diabetologia.
The researchers conducted a systematic search in PubMed MEDLINE to identify reports on the links between diabetes and cancer. Additional reports were identified from the reference lists of the relevant studies.
Only those cohort studies providing relative risks (RRs) for the association between diabetes and cancer for both women and men were included. In total, 107 relevant articles were identified, along with 36 cohorts of individual participant data.
RRs for cancer were obtained for patients with diabetes (types 1 and 2 combined) versus those without diabetes, for both men and women. The women-to-men ratios of these relative risk ratios (RRRs) were then calculated to determine the excess risk in women if present.
Data on all-site cancer was available from 47 studies, involving 121 cohorts and 19,239,302 individuals.
Diabetics vs non-diabetics
Women with diabetes had a 27% higher risk of all-site cancer compared to women without diabetes (RR=1.27; 95% CI 1.21, 1.32; P<0.001).
For men, the risk of all-site cancer was 19% higher among those with diabetes than those without (RR=1.19; 95% CI 1.13, 1.25; P<0.001).
There were several hematologic malignancies for which diabetics had an increased risk, as shown in the following table.
Cancer type | RR for women (99% CI) | RR for men (99% CI) |
Lymphatic and hematopoietic tissue | 1.24 (1.05, 1.46)* | 1.21 (0.98, 1.48) |
Leukemia | 1.53 (1.00, 2.33) | 1.22 (0.80, 1.85) |
Myeloid leukemia | 0.83 (0.39, 1.76) | 1.12 (0.77, 1.62) |
Acute myeloid leukemia | 1.33 (1.12, 1.57)* | 1.14 (0.56, 2.33) |
Chronic myeloid leukemia | 1.67 (1.27, 2.20)* | 1.62 (1.32, 1.98)* |
Lymphoid leukemia | 1.74 (0.31, 9.79) | 1.20 (0.86, 1.68) |
Lymphoma | 2.31 (0.57, 9.30) | 1.80 (0.68, 4.75) |
Non-Hodgkin lymphoma | 1.16 (1.02, 1.32)* | 1.20 (1.08, 1.34)* |
Hodgkin lymphoma | 1.20 (0.61, 2.38) | 1.36 (1.05, 1.77)* |
Multiple myeloma | 1.19 (0.97, 1.47) | 1.12 (0.90, 1.41) |
*denotes statistical significance with a P value < 0.01 |
Sex comparison
Calculation of the women-to-men ratio revealed that women with diabetes had a 6% greater excess risk of all-site cancer compared to men with diabetes (RRR=1.06; 95% CI 1.03, 1.09; P<0.001).
The women-to-men ratios also showed significantly higher risks for female diabetics for:
- Kidney cancer—RRR=1.11 (99% CI 1.04, 1.18; P<0.001)
- Oral cancer—RRR=1.13 (99% CI 1.00, 1.28; P=0.009)
- Stomach cancer—RRR=1.14 (99% CI 1.07, 1.22; P<0.001)
- Leukemia—RRR=1.15 (99% CI 1.02, 1.28; P=0.002).
However, women had a significantly lower risk of liver cancer (RRR=0.88; 99% CI 0.79, 0.99; P=0.005).
There are several possible reasons for the excess cancer risk observed in women, according to study author Sanne Peters, PhD, of The George Institute for Global Health at the University of Oxford in the UK.
For example, on average, women are in the pre-diabetic state of impaired glucose tolerance 2 years longer than men.
“Historically, we know that women are often under-treated when they first present with symptoms of diabetes, are less likely to receive intensive care, and are not taking the same levels of medications as men,” Dr Peters said. “All of these could go some way into explaining why women are at greater risk of developing cancer, but, without more research, we can’t be certain.”
A review of data from more than 19 million people indicates that diabetes significantly raises a person’s risk of developing cancer.
When researchers compared patients with diabetes and without, both male and female diabetics had an increased risk of leukemias and lymphomas as well as certain solid tumors.
Researchers also found that diabetes conferred a higher cancer risk for women than men, both for all cancers combined and for some specific cancers, including leukemia.
“The link between diabetes and the risk of developing cancer is now firmly established,” said Toshiaki Ohkuma, PhD, of The George Institute for Global Health at the University of New South Wales in Australia.
“We have also demonstrated, for the first time, that women with diabetes are more likely to develop any form of cancer and have a significantly higher chance of developing kidney, oral, and stomach cancers and leukemia.”
Dr Ohkuma and his colleagues reported these findings in Diabetologia.
The researchers conducted a systematic search in PubMed MEDLINE to identify reports on the links between diabetes and cancer. Additional reports were identified from the reference lists of the relevant studies.
Only those cohort studies providing relative risks (RRs) for the association between diabetes and cancer for both women and men were included. In total, 107 relevant articles were identified, along with 36 cohorts of individual participant data.
RRs for cancer were obtained for patients with diabetes (types 1 and 2 combined) versus those without diabetes, for both men and women. The women-to-men ratios of these relative risk ratios (RRRs) were then calculated to determine the excess risk in women if present.
Data on all-site cancer was available from 47 studies, involving 121 cohorts and 19,239,302 individuals.
Diabetics vs non-diabetics
Women with diabetes had a 27% higher risk of all-site cancer compared to women without diabetes (RR=1.27; 95% CI 1.21, 1.32; P<0.001).
For men, the risk of all-site cancer was 19% higher among those with diabetes than those without (RR=1.19; 95% CI 1.13, 1.25; P<0.001).
There were several hematologic malignancies for which diabetics had an increased risk, as shown in the following table.
Cancer type | RR for women (99% CI) | RR for men (99% CI) |
Lymphatic and hematopoietic tissue | 1.24 (1.05, 1.46)* | 1.21 (0.98, 1.48) |
Leukemia | 1.53 (1.00, 2.33) | 1.22 (0.80, 1.85) |
Myeloid leukemia | 0.83 (0.39, 1.76) | 1.12 (0.77, 1.62) |
Acute myeloid leukemia | 1.33 (1.12, 1.57)* | 1.14 (0.56, 2.33) |
Chronic myeloid leukemia | 1.67 (1.27, 2.20)* | 1.62 (1.32, 1.98)* |
Lymphoid leukemia | 1.74 (0.31, 9.79) | 1.20 (0.86, 1.68) |
Lymphoma | 2.31 (0.57, 9.30) | 1.80 (0.68, 4.75) |
Non-Hodgkin lymphoma | 1.16 (1.02, 1.32)* | 1.20 (1.08, 1.34)* |
Hodgkin lymphoma | 1.20 (0.61, 2.38) | 1.36 (1.05, 1.77)* |
Multiple myeloma | 1.19 (0.97, 1.47) | 1.12 (0.90, 1.41) |
*denotes statistical significance with a P value < 0.01 |
Sex comparison
Calculation of the women-to-men ratio revealed that women with diabetes had a 6% greater excess risk of all-site cancer compared to men with diabetes (RRR=1.06; 95% CI 1.03, 1.09; P<0.001).
The women-to-men ratios also showed significantly higher risks for female diabetics for:
- Kidney cancer—RRR=1.11 (99% CI 1.04, 1.18; P<0.001)
- Oral cancer—RRR=1.13 (99% CI 1.00, 1.28; P=0.009)
- Stomach cancer—RRR=1.14 (99% CI 1.07, 1.22; P<0.001)
- Leukemia—RRR=1.15 (99% CI 1.02, 1.28; P=0.002).
However, women had a significantly lower risk of liver cancer (RRR=0.88; 99% CI 0.79, 0.99; P=0.005).
There are several possible reasons for the excess cancer risk observed in women, according to study author Sanne Peters, PhD, of The George Institute for Global Health at the University of Oxford in the UK.
For example, on average, women are in the pre-diabetic state of impaired glucose tolerance 2 years longer than men.
“Historically, we know that women are often under-treated when they first present with symptoms of diabetes, are less likely to receive intensive care, and are not taking the same levels of medications as men,” Dr Peters said. “All of these could go some way into explaining why women are at greater risk of developing cancer, but, without more research, we can’t be certain.”
Study suggests dasatinib could treat AML, JMML
New research suggests dasatinib could treat certain patients with juvenile myelomonocytic leukemia (JMML) or acute myeloid leukemia (AML).
The study showed that TNK2 inhibition has a negative effect on PTPN11-mutant leukemias.
PTPN11-mutant JMML and AML cells were sensitive to treatment with dasatinib, which inhibits TNK2.
Dasatinib also induced hematologic remission in a patient with PTPN11-mutant JMML.
Investigators reported these results in Science Signaling.
Past research showed that mutations in PTPN11 result in excessive cell proliferation and drive tumor growth in some cases of JMML and AML.
In the current study, investigators analyzed PTPN11-mutated leukemia cells and found that PTPN11 is activated by TNK2.
The investigators said TNK2 phosphorylates PTPN11, which then dephosphorylates TNK2 in a negative feedback loop. They also found that coexpression of TNK2 and mutant PTPN11 results in “robust” MAPK pathway activation.
Inhibiting TNK2 with dasatinib blocked MAPK signaling and colony formation in vitro.
Additional experiments showed that PTPN11-mutant AML samples were significantly more sensitive to dasatinib than wild-type AML samples.
Investigators also tested dasatinib in a sample from a JMML patient carrying a PTPN11 G60R mutation.
This patient’s cells were 10 times more sensitive to dasatinib than the average sample from a cohort of 151 patients who had AML, acute lymphoblastic leukemia, myeloproliferative neoplasms, or chronic lymphocytic leukemia.
Because the JMML patient’s cells were so responsive to dasatinib, the investigators decided to administer the drug to the patient.
The patient achieved sustained hematologic remission with dasatinib, and this allowed him to receive a stem cell transplant using an unrelated cord blood donor. The patient had previously failed 2 transplants (with myeloablative conditioning) from a matched sibling donor.
The third transplant prolonged the patient’s life by a year, but he eventually died of relapsed disease.
The investigators said this case study and the in vitro results support further investigation into the efficacy of dasatinib and other TNK2 inhibitors in PTPN11-mutant leukemias.
New research suggests dasatinib could treat certain patients with juvenile myelomonocytic leukemia (JMML) or acute myeloid leukemia (AML).
The study showed that TNK2 inhibition has a negative effect on PTPN11-mutant leukemias.
PTPN11-mutant JMML and AML cells were sensitive to treatment with dasatinib, which inhibits TNK2.
Dasatinib also induced hematologic remission in a patient with PTPN11-mutant JMML.
Investigators reported these results in Science Signaling.
Past research showed that mutations in PTPN11 result in excessive cell proliferation and drive tumor growth in some cases of JMML and AML.
In the current study, investigators analyzed PTPN11-mutated leukemia cells and found that PTPN11 is activated by TNK2.
The investigators said TNK2 phosphorylates PTPN11, which then dephosphorylates TNK2 in a negative feedback loop. They also found that coexpression of TNK2 and mutant PTPN11 results in “robust” MAPK pathway activation.
Inhibiting TNK2 with dasatinib blocked MAPK signaling and colony formation in vitro.
Additional experiments showed that PTPN11-mutant AML samples were significantly more sensitive to dasatinib than wild-type AML samples.
Investigators also tested dasatinib in a sample from a JMML patient carrying a PTPN11 G60R mutation.
This patient’s cells were 10 times more sensitive to dasatinib than the average sample from a cohort of 151 patients who had AML, acute lymphoblastic leukemia, myeloproliferative neoplasms, or chronic lymphocytic leukemia.
Because the JMML patient’s cells were so responsive to dasatinib, the investigators decided to administer the drug to the patient.
The patient achieved sustained hematologic remission with dasatinib, and this allowed him to receive a stem cell transplant using an unrelated cord blood donor. The patient had previously failed 2 transplants (with myeloablative conditioning) from a matched sibling donor.
The third transplant prolonged the patient’s life by a year, but he eventually died of relapsed disease.
The investigators said this case study and the in vitro results support further investigation into the efficacy of dasatinib and other TNK2 inhibitors in PTPN11-mutant leukemias.
New research suggests dasatinib could treat certain patients with juvenile myelomonocytic leukemia (JMML) or acute myeloid leukemia (AML).
The study showed that TNK2 inhibition has a negative effect on PTPN11-mutant leukemias.
PTPN11-mutant JMML and AML cells were sensitive to treatment with dasatinib, which inhibits TNK2.
Dasatinib also induced hematologic remission in a patient with PTPN11-mutant JMML.
Investigators reported these results in Science Signaling.
Past research showed that mutations in PTPN11 result in excessive cell proliferation and drive tumor growth in some cases of JMML and AML.
In the current study, investigators analyzed PTPN11-mutated leukemia cells and found that PTPN11 is activated by TNK2.
The investigators said TNK2 phosphorylates PTPN11, which then dephosphorylates TNK2 in a negative feedback loop. They also found that coexpression of TNK2 and mutant PTPN11 results in “robust” MAPK pathway activation.
Inhibiting TNK2 with dasatinib blocked MAPK signaling and colony formation in vitro.
Additional experiments showed that PTPN11-mutant AML samples were significantly more sensitive to dasatinib than wild-type AML samples.
Investigators also tested dasatinib in a sample from a JMML patient carrying a PTPN11 G60R mutation.
This patient’s cells were 10 times more sensitive to dasatinib than the average sample from a cohort of 151 patients who had AML, acute lymphoblastic leukemia, myeloproliferative neoplasms, or chronic lymphocytic leukemia.
Because the JMML patient’s cells were so responsive to dasatinib, the investigators decided to administer the drug to the patient.
The patient achieved sustained hematologic remission with dasatinib, and this allowed him to receive a stem cell transplant using an unrelated cord blood donor. The patient had previously failed 2 transplants (with myeloablative conditioning) from a matched sibling donor.
The third transplant prolonged the patient’s life by a year, but he eventually died of relapsed disease.
The investigators said this case study and the in vitro results support further investigation into the efficacy of dasatinib and other TNK2 inhibitors in PTPN11-mutant leukemias.
Explaining enasidenib resistance in AML
New research helps explain enasidenib resistance among patients with IDH2-mutant acute myeloid leukemia (AML).
Researchers found that leukemic cells stop responding to enasidenib when IDH2 clones develop additional mutations.
This may mean that enasidenib will have to be combined with other drugs to prevent AML relapse, the researchers said.
They reported their findings in Nature Medicine.
Previous research indicated that enasidenib prompts differentiation to induce responses in AML. In a phase 1/2 trial, enasidenib produced responses in about 40% of patients with relapsed/refractory, IDH2-mutated AML. However, most patients eventually relapsed.
“[T]he initial studies did not show which AML cells responded to enasidenib and started to differentiate again,” said Stéphane de Botton, MD, PhD, of Institut Gustave Roussy in Villejuif, France.
“It was also unclear how the cells become resistant to therapy. We wanted to answer these questions.”
To do so, Dr de Botton and his colleagues analyzed sequential samples from 37 AML patients treated with enasidenib on the phase 1/2 trial. Thirty of these patients had initially responded to the drug.
“We used techniques to study genetic mutations on a cell-by-cell basis and reconstructed the ‘family tree’ of a patient’s AML,” said Lynn Quek, MD, of the University of Oxford in the UK.
“We then tracked changes in the family of AML cells as they responded to enasidenib and as patients lost response to the drug. This is the first time that anyone has done such a detailed study at a single-cell level.”
The researchers said they observed variable differentiation arrest in IDH2-mutant clones before enasidenib treatment.
Overall, treatment promoted hematopoietic differentiation from either terminal or ancestral mutant clones. However, enasidenib also promoted differentiation of nonmutant cells in a minority of patients.
When the researchers compared samples taken at diagnosis and relapse, they did not find second-site mutations in IDH2 at relapse.
The team said relapse was the result of clonal evolution or selection of terminal or ancestral clones, which suggests there are multiple pathways that could potentially be targeted to restore differentiation arrest.
“We have provided genetic proof that enasidenib was able to differentiate cancer cells so that some of their normal functions were restored, even though they still contained the IDH2 mutation,” said Virginie Penard-Lacronique, of Gustave Roussy.
“This is important because, unless we can track these clones, we don’t know whether the mature cells in a patient are coming from normal cells after all the cancer cells have been killed or from leukemic cells that are now able to mature. In this paper, we show that, in 4 out of 5 cases, the mature cells are coming from leukemic bone marrow cells that can be induced to differentiate by this new drug.”
The researchers said these results suggest enasidenib must be combined with other drugs to prevent AML relapse.
“Now that we have shown that [enasidenib] needs to be combined with other drugs to stop the cancer returning, we think that it’s important that the combination therapy should be given to AML patients early on in their disease,” Dr de Botton said. “International trials are now taking place comparing combinations of enasidenib and other drugs with the normal standard of care.”
New research helps explain enasidenib resistance among patients with IDH2-mutant acute myeloid leukemia (AML).
Researchers found that leukemic cells stop responding to enasidenib when IDH2 clones develop additional mutations.
This may mean that enasidenib will have to be combined with other drugs to prevent AML relapse, the researchers said.
They reported their findings in Nature Medicine.
Previous research indicated that enasidenib prompts differentiation to induce responses in AML. In a phase 1/2 trial, enasidenib produced responses in about 40% of patients with relapsed/refractory, IDH2-mutated AML. However, most patients eventually relapsed.
“[T]he initial studies did not show which AML cells responded to enasidenib and started to differentiate again,” said Stéphane de Botton, MD, PhD, of Institut Gustave Roussy in Villejuif, France.
“It was also unclear how the cells become resistant to therapy. We wanted to answer these questions.”
To do so, Dr de Botton and his colleagues analyzed sequential samples from 37 AML patients treated with enasidenib on the phase 1/2 trial. Thirty of these patients had initially responded to the drug.
“We used techniques to study genetic mutations on a cell-by-cell basis and reconstructed the ‘family tree’ of a patient’s AML,” said Lynn Quek, MD, of the University of Oxford in the UK.
“We then tracked changes in the family of AML cells as they responded to enasidenib and as patients lost response to the drug. This is the first time that anyone has done such a detailed study at a single-cell level.”
The researchers said they observed variable differentiation arrest in IDH2-mutant clones before enasidenib treatment.
Overall, treatment promoted hematopoietic differentiation from either terminal or ancestral mutant clones. However, enasidenib also promoted differentiation of nonmutant cells in a minority of patients.
When the researchers compared samples taken at diagnosis and relapse, they did not find second-site mutations in IDH2 at relapse.
The team said relapse was the result of clonal evolution or selection of terminal or ancestral clones, which suggests there are multiple pathways that could potentially be targeted to restore differentiation arrest.
“We have provided genetic proof that enasidenib was able to differentiate cancer cells so that some of their normal functions were restored, even though they still contained the IDH2 mutation,” said Virginie Penard-Lacronique, of Gustave Roussy.
“This is important because, unless we can track these clones, we don’t know whether the mature cells in a patient are coming from normal cells after all the cancer cells have been killed or from leukemic cells that are now able to mature. In this paper, we show that, in 4 out of 5 cases, the mature cells are coming from leukemic bone marrow cells that can be induced to differentiate by this new drug.”
The researchers said these results suggest enasidenib must be combined with other drugs to prevent AML relapse.
“Now that we have shown that [enasidenib] needs to be combined with other drugs to stop the cancer returning, we think that it’s important that the combination therapy should be given to AML patients early on in their disease,” Dr de Botton said. “International trials are now taking place comparing combinations of enasidenib and other drugs with the normal standard of care.”
New research helps explain enasidenib resistance among patients with IDH2-mutant acute myeloid leukemia (AML).
Researchers found that leukemic cells stop responding to enasidenib when IDH2 clones develop additional mutations.
This may mean that enasidenib will have to be combined with other drugs to prevent AML relapse, the researchers said.
They reported their findings in Nature Medicine.
Previous research indicated that enasidenib prompts differentiation to induce responses in AML. In a phase 1/2 trial, enasidenib produced responses in about 40% of patients with relapsed/refractory, IDH2-mutated AML. However, most patients eventually relapsed.
“[T]he initial studies did not show which AML cells responded to enasidenib and started to differentiate again,” said Stéphane de Botton, MD, PhD, of Institut Gustave Roussy in Villejuif, France.
“It was also unclear how the cells become resistant to therapy. We wanted to answer these questions.”
To do so, Dr de Botton and his colleagues analyzed sequential samples from 37 AML patients treated with enasidenib on the phase 1/2 trial. Thirty of these patients had initially responded to the drug.
“We used techniques to study genetic mutations on a cell-by-cell basis and reconstructed the ‘family tree’ of a patient’s AML,” said Lynn Quek, MD, of the University of Oxford in the UK.
“We then tracked changes in the family of AML cells as they responded to enasidenib and as patients lost response to the drug. This is the first time that anyone has done such a detailed study at a single-cell level.”
The researchers said they observed variable differentiation arrest in IDH2-mutant clones before enasidenib treatment.
Overall, treatment promoted hematopoietic differentiation from either terminal or ancestral mutant clones. However, enasidenib also promoted differentiation of nonmutant cells in a minority of patients.
When the researchers compared samples taken at diagnosis and relapse, they did not find second-site mutations in IDH2 at relapse.
The team said relapse was the result of clonal evolution or selection of terminal or ancestral clones, which suggests there are multiple pathways that could potentially be targeted to restore differentiation arrest.
“We have provided genetic proof that enasidenib was able to differentiate cancer cells so that some of their normal functions were restored, even though they still contained the IDH2 mutation,” said Virginie Penard-Lacronique, of Gustave Roussy.
“This is important because, unless we can track these clones, we don’t know whether the mature cells in a patient are coming from normal cells after all the cancer cells have been killed or from leukemic cells that are now able to mature. In this paper, we show that, in 4 out of 5 cases, the mature cells are coming from leukemic bone marrow cells that can be induced to differentiate by this new drug.”
The researchers said these results suggest enasidenib must be combined with other drugs to prevent AML relapse.
“Now that we have shown that [enasidenib] needs to be combined with other drugs to stop the cancer returning, we think that it’s important that the combination therapy should be given to AML patients early on in their disease,” Dr de Botton said. “International trials are now taking place comparing combinations of enasidenib and other drugs with the normal standard of care.”
CAR T Therapy: From Bench to Bedside and Back
Release Date: July 15, 2018
Expiration Date: July 14, 2019
Note: This activity is no longer available for credit
Introductory Comments: (Duration: 9 minutes)
Aaron P. Rapoport, MD
Bone Marrow Transplant Program
University of Maryland School of Medicine
Baltimore, MD
Presentation: (Duration: 39 minutes)
Carl H. June, MD
Richard W. Vague Professor in Immunotherapy
Perelman School of Medicine
University of Pennsylvania
Philadelphia, PA
Provided by:
Learning Objectives
• Review clinical data and individual case studies to determine where CAR T-cell therapy might be appropriate in the treatment of adult and pediatric patients with leukemia, lymphoma, and multiple myeloma.
• Discuss the management of cytotoxicity of CAR T-cell therapy.
Target Audience
Hematologists, oncologists, and other members of the healthcare team who treat or manage patients with hematologic malignancies.
Statement of Need
It is critical that clinicians managing patients with acute leukemia and other hematologic malignancies are cognizant of exciting breakthroughs and are also able to integrate recent progress into practice. However, given the overwhelming influx of data, it is no surprise that many hematology professionals face difficulties in identifying the most relevant findings for clinical practice. Hematologists are unable to stay abreast of the latest evidence on investigational agents. Educational programs are thus crucial to address this important professional practice gap.
Faculty
Carl H. June, MD
Richard W. Vague Professor in Immunotherapy
Perelman School of Medicine
University of Pennsylvania
Philadelphia, PA
Disclosures: Consultant: Novartis; Grant/Research support and royalties/IPR: Novartis
Stockholder: Tmunity Therapeutics, Inc.
Aaron P. Rapoport, MD
Bone Marrow Transplant Program
University of Maryland School of Medicine
Baltimore, Maryland
Disclosures: No relevant financial relationships with a commercial supporter
Permissions
- Slide 3: Complex tumor, host and environmental factors govern the strength and timing of anti-cancer immune responses
- Reprinted from Immunity, Vol 39/No 1, Chen DS, Mellman I, Oncology meets immunology: the cancer-immunity cycle, pp 1-10, 2013, with permission from Elsevier
- Slide 9: Genes differentially expressed in CART19 cellular infusion products from CLL patients
- From Fraietta JA, Lacey SF, Orlando EJ, . . . June CH, Melenhorst JJ. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med 2018; 24:563-571
- Slide 10: Characterization of CLL CAR T cells in NSG CLL model
- Same as slide 9
- Slide 15: First adult ALL patient
- Photos originally published in Kaiser Health News/Photo courtesy of Dr Keith Eaton. Available at: https://khn.org/news/cascade-of-costs-could-push-new-gene-therapy-above-1-million-per-patient/
- Slide 21: Efficient trafficking of CTL019 T Cells to CNS in ALL
- From N Engl J Med, Grupp SA, Kalos M, Barrett D, . . V. June CH, Chimeric antigen receptor-modified T cells for acute lymphoid leukemia, Volume No 368, pp 1509-1518. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission from Massachusetts Medical Society.
- Slide 26: Long-term persistence and expression of CTL019 is associated with durable remission in leukemia: Predictive Biomarker
- From Porter DL, Hwang WT, Frey NV . . . June CH. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med 2015; 7(303):303ra139. Reprinted with permission from AAAS.
- Slide 28: Rapid massive expansion of clonal CART cell population in patient #10
- Initially published in Fraietta JA, Nobles CL, Sammons MA, . . . June CH, Melenhors JJ. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 2018; 558(7709):307-312
- Slide 29: Mapping CAR integration site in Pt #10
- Same as slide 28.
- Slide 31: Long-term stable persistence of TET2-deficient CAR T cells in Pt #10
- Same as slide 28
- Slide 32: Epigenetic and genetic changes uncovered by ATAC-seq in Pt #10
- Same as slide 28.
- Slide 33: TET2 knock down in healthy donor T cells
- Same as slide 28.
- Slide 34: TET2 knock down in healthy donor T cells
- Same as slide 28.
- Slide 36: CAR T for myeloma: BCMA
- From Rickert RC, Jellusova J, Miletic AV. Signaling by the tumor necrosis factor receptor superfamily in B-cell biology and disease. Immunol Rev 2011; 244(1):115-33. Reprinted with permission from John Wiley and Sons.
- Slide 38: CAR T for myeloma: Patient #1
- Photo originally published by UT Southwestern Medical Center. Available at: https://www.utsouthwestern.edu/newsroom/articles/year-2018/wright-car-t.html
- Slide 39: Autoimmunity is the “Achilles’ Heel” of immunotherapy
- First published in June CH, Warshauer JT, and Bluestone JA. Is autoimmunity the Achilles’ heel of cancer immunotherapy? Nat Med 2017;23(5):540-7
- Slide 41: Multiplex CRISPR /Cas9 editing: Universal T cells TCR, HLA, PD-1, CTLA-4 and Fas
- From Ren J, Zhang X, Liu X, Fang C, Jiang S, June CH, Zhao Y. A versatile system for rapid multiplex genome-edited CAR T cell generation. Oncotarget 2017; 8:17002-17011.
- Slide 45: CAR T-cell trials for cancer are now global
- From June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science 2018; 359:1361-1365. Reprinted with permission from AAAS.
Disclaimer
The content and views presented in this educational activity are those of the author and do not necessarily reflect those of Hemedicus or Frontline Medical Communications. This material is prepared based upon a review of multiple sources of information, but it is not exhaustive of the subject matter. Therefore, healthcare professionals and other individuals should review and consider other publications and materials on the subject matter before relying solely upon the information contained within this educational activity.
Release Date: July 15, 2018
Expiration Date: July 14, 2019
Note: This activity is no longer available for credit
Introductory Comments: (Duration: 9 minutes)
Aaron P. Rapoport, MD
Bone Marrow Transplant Program
University of Maryland School of Medicine
Baltimore, MD
Presentation: (Duration: 39 minutes)
Carl H. June, MD
Richard W. Vague Professor in Immunotherapy
Perelman School of Medicine
University of Pennsylvania
Philadelphia, PA
Provided by:
Learning Objectives
• Review clinical data and individual case studies to determine where CAR T-cell therapy might be appropriate in the treatment of adult and pediatric patients with leukemia, lymphoma, and multiple myeloma.
• Discuss the management of cytotoxicity of CAR T-cell therapy.
Target Audience
Hematologists, oncologists, and other members of the healthcare team who treat or manage patients with hematologic malignancies.
Statement of Need
It is critical that clinicians managing patients with acute leukemia and other hematologic malignancies are cognizant of exciting breakthroughs and are also able to integrate recent progress into practice. However, given the overwhelming influx of data, it is no surprise that many hematology professionals face difficulties in identifying the most relevant findings for clinical practice. Hematologists are unable to stay abreast of the latest evidence on investigational agents. Educational programs are thus crucial to address this important professional practice gap.
Faculty
Carl H. June, MD
Richard W. Vague Professor in Immunotherapy
Perelman School of Medicine
University of Pennsylvania
Philadelphia, PA
Disclosures: Consultant: Novartis; Grant/Research support and royalties/IPR: Novartis
Stockholder: Tmunity Therapeutics, Inc.
Aaron P. Rapoport, MD
Bone Marrow Transplant Program
University of Maryland School of Medicine
Baltimore, Maryland
Disclosures: No relevant financial relationships with a commercial supporter
Permissions
- Slide 3: Complex tumor, host and environmental factors govern the strength and timing of anti-cancer immune responses
- Reprinted from Immunity, Vol 39/No 1, Chen DS, Mellman I, Oncology meets immunology: the cancer-immunity cycle, pp 1-10, 2013, with permission from Elsevier
- Slide 9: Genes differentially expressed in CART19 cellular infusion products from CLL patients
- From Fraietta JA, Lacey SF, Orlando EJ, . . . June CH, Melenhorst JJ. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med 2018; 24:563-571
- Slide 10: Characterization of CLL CAR T cells in NSG CLL model
- Same as slide 9
- Slide 15: First adult ALL patient
- Photos originally published in Kaiser Health News/Photo courtesy of Dr Keith Eaton. Available at: https://khn.org/news/cascade-of-costs-could-push-new-gene-therapy-above-1-million-per-patient/
- Slide 21: Efficient trafficking of CTL019 T Cells to CNS in ALL
- From N Engl J Med, Grupp SA, Kalos M, Barrett D, . . V. June CH, Chimeric antigen receptor-modified T cells for acute lymphoid leukemia, Volume No 368, pp 1509-1518. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission from Massachusetts Medical Society.
- Slide 26: Long-term persistence and expression of CTL019 is associated with durable remission in leukemia: Predictive Biomarker
- From Porter DL, Hwang WT, Frey NV . . . June CH. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med 2015; 7(303):303ra139. Reprinted with permission from AAAS.
- Slide 28: Rapid massive expansion of clonal CART cell population in patient #10
- Initially published in Fraietta JA, Nobles CL, Sammons MA, . . . June CH, Melenhors JJ. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 2018; 558(7709):307-312
- Slide 29: Mapping CAR integration site in Pt #10
- Same as slide 28.
- Slide 31: Long-term stable persistence of TET2-deficient CAR T cells in Pt #10
- Same as slide 28
- Slide 32: Epigenetic and genetic changes uncovered by ATAC-seq in Pt #10
- Same as slide 28.
- Slide 33: TET2 knock down in healthy donor T cells
- Same as slide 28.
- Slide 34: TET2 knock down in healthy donor T cells
- Same as slide 28.
- Slide 36: CAR T for myeloma: BCMA
- From Rickert RC, Jellusova J, Miletic AV. Signaling by the tumor necrosis factor receptor superfamily in B-cell biology and disease. Immunol Rev 2011; 244(1):115-33. Reprinted with permission from John Wiley and Sons.
- Slide 38: CAR T for myeloma: Patient #1
- Photo originally published by UT Southwestern Medical Center. Available at: https://www.utsouthwestern.edu/newsroom/articles/year-2018/wright-car-t.html
- Slide 39: Autoimmunity is the “Achilles’ Heel” of immunotherapy
- First published in June CH, Warshauer JT, and Bluestone JA. Is autoimmunity the Achilles’ heel of cancer immunotherapy? Nat Med 2017;23(5):540-7
- Slide 41: Multiplex CRISPR /Cas9 editing: Universal T cells TCR, HLA, PD-1, CTLA-4 and Fas
- From Ren J, Zhang X, Liu X, Fang C, Jiang S, June CH, Zhao Y. A versatile system for rapid multiplex genome-edited CAR T cell generation. Oncotarget 2017; 8:17002-17011.
- Slide 45: CAR T-cell trials for cancer are now global
- From June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science 2018; 359:1361-1365. Reprinted with permission from AAAS.
Disclaimer
The content and views presented in this educational activity are those of the author and do not necessarily reflect those of Hemedicus or Frontline Medical Communications. This material is prepared based upon a review of multiple sources of information, but it is not exhaustive of the subject matter. Therefore, healthcare professionals and other individuals should review and consider other publications and materials on the subject matter before relying solely upon the information contained within this educational activity.
Release Date: July 15, 2018
Expiration Date: July 14, 2019
Note: This activity is no longer available for credit
Introductory Comments: (Duration: 9 minutes)
Aaron P. Rapoport, MD
Bone Marrow Transplant Program
University of Maryland School of Medicine
Baltimore, MD
Presentation: (Duration: 39 minutes)
Carl H. June, MD
Richard W. Vague Professor in Immunotherapy
Perelman School of Medicine
University of Pennsylvania
Philadelphia, PA
Provided by:
Learning Objectives
• Review clinical data and individual case studies to determine where CAR T-cell therapy might be appropriate in the treatment of adult and pediatric patients with leukemia, lymphoma, and multiple myeloma.
• Discuss the management of cytotoxicity of CAR T-cell therapy.
Target Audience
Hematologists, oncologists, and other members of the healthcare team who treat or manage patients with hematologic malignancies.
Statement of Need
It is critical that clinicians managing patients with acute leukemia and other hematologic malignancies are cognizant of exciting breakthroughs and are also able to integrate recent progress into practice. However, given the overwhelming influx of data, it is no surprise that many hematology professionals face difficulties in identifying the most relevant findings for clinical practice. Hematologists are unable to stay abreast of the latest evidence on investigational agents. Educational programs are thus crucial to address this important professional practice gap.
Faculty
Carl H. June, MD
Richard W. Vague Professor in Immunotherapy
Perelman School of Medicine
University of Pennsylvania
Philadelphia, PA
Disclosures: Consultant: Novartis; Grant/Research support and royalties/IPR: Novartis
Stockholder: Tmunity Therapeutics, Inc.
Aaron P. Rapoport, MD
Bone Marrow Transplant Program
University of Maryland School of Medicine
Baltimore, Maryland
Disclosures: No relevant financial relationships with a commercial supporter
Permissions
- Slide 3: Complex tumor, host and environmental factors govern the strength and timing of anti-cancer immune responses
- Reprinted from Immunity, Vol 39/No 1, Chen DS, Mellman I, Oncology meets immunology: the cancer-immunity cycle, pp 1-10, 2013, with permission from Elsevier
- Slide 9: Genes differentially expressed in CART19 cellular infusion products from CLL patients
- From Fraietta JA, Lacey SF, Orlando EJ, . . . June CH, Melenhorst JJ. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med 2018; 24:563-571
- Slide 10: Characterization of CLL CAR T cells in NSG CLL model
- Same as slide 9
- Slide 15: First adult ALL patient
- Photos originally published in Kaiser Health News/Photo courtesy of Dr Keith Eaton. Available at: https://khn.org/news/cascade-of-costs-could-push-new-gene-therapy-above-1-million-per-patient/
- Slide 21: Efficient trafficking of CTL019 T Cells to CNS in ALL
- From N Engl J Med, Grupp SA, Kalos M, Barrett D, . . V. June CH, Chimeric antigen receptor-modified T cells for acute lymphoid leukemia, Volume No 368, pp 1509-1518. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission from Massachusetts Medical Society.
- Slide 26: Long-term persistence and expression of CTL019 is associated with durable remission in leukemia: Predictive Biomarker
- From Porter DL, Hwang WT, Frey NV . . . June CH. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med 2015; 7(303):303ra139. Reprinted with permission from AAAS.
- Slide 28: Rapid massive expansion of clonal CART cell population in patient #10
- Initially published in Fraietta JA, Nobles CL, Sammons MA, . . . June CH, Melenhors JJ. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 2018; 558(7709):307-312
- Slide 29: Mapping CAR integration site in Pt #10
- Same as slide 28.
- Slide 31: Long-term stable persistence of TET2-deficient CAR T cells in Pt #10
- Same as slide 28
- Slide 32: Epigenetic and genetic changes uncovered by ATAC-seq in Pt #10
- Same as slide 28.
- Slide 33: TET2 knock down in healthy donor T cells
- Same as slide 28.
- Slide 34: TET2 knock down in healthy donor T cells
- Same as slide 28.
- Slide 36: CAR T for myeloma: BCMA
- From Rickert RC, Jellusova J, Miletic AV. Signaling by the tumor necrosis factor receptor superfamily in B-cell biology and disease. Immunol Rev 2011; 244(1):115-33. Reprinted with permission from John Wiley and Sons.
- Slide 38: CAR T for myeloma: Patient #1
- Photo originally published by UT Southwestern Medical Center. Available at: https://www.utsouthwestern.edu/newsroom/articles/year-2018/wright-car-t.html
- Slide 39: Autoimmunity is the “Achilles’ Heel” of immunotherapy
- First published in June CH, Warshauer JT, and Bluestone JA. Is autoimmunity the Achilles’ heel of cancer immunotherapy? Nat Med 2017;23(5):540-7
- Slide 41: Multiplex CRISPR /Cas9 editing: Universal T cells TCR, HLA, PD-1, CTLA-4 and Fas
- From Ren J, Zhang X, Liu X, Fang C, Jiang S, June CH, Zhao Y. A versatile system for rapid multiplex genome-edited CAR T cell generation. Oncotarget 2017; 8:17002-17011.
- Slide 45: CAR T-cell trials for cancer are now global
- From June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science 2018; 359:1361-1365. Reprinted with permission from AAAS.
Disclaimer
The content and views presented in this educational activity are those of the author and do not necessarily reflect those of Hemedicus or Frontline Medical Communications. This material is prepared based upon a review of multiple sources of information, but it is not exhaustive of the subject matter. Therefore, healthcare professionals and other individuals should review and consider other publications and materials on the subject matter before relying solely upon the information contained within this educational activity.
Genentech submits sNDA for venetoclax in untreated AML
A supplemental new drug application (sNDA) for venetoclax (Venclexta) used in combination with either a hypomethylating agent or low-dose cytarabine (LDAC) for previously untreated acute myeloid leukemia has been submitted to the Food and Drug Administration by Genentech, which developed it.
Specifically, the sNDA is for these drug combinations in the treatment of AML patients ineligible for intensive chemotherapy, according to the announcement from Genentech.
The sNDA is based on results of two trials that included patients in this population. In the phase 1b M14-358 (NCT02203773), venetoclax was combined with either azacitidine or decitabine; patients treated with 400 mg of venetoclax had a complete remission rate of 73%, and the median overall survival across all doses of venetoclax was 17.5 months. Low white blood cell count with fever, low white blood cell count, anemia, low platelet count, and decreased potassium levels were the most common grade 3/4 adverse events (occurring in 10% or more of patients). In the phase 1b/2 study M14-387 (NCT02287233), venetoclax was used in combination with LDAC; patients treated with a 600-mg dose of venetoclax showed a complete response rate of 62%, and a median overall survival of 11.4 months. Low white blood cell count with fever, decreased potassium levels, pneumonia, disease progression, decreased phosphate levels, high blood pressure, and sepsis were the most common grade 3/4 adverse events seen in this study.
This sNDA follows FDA breakthrough therapy designations, based on these same trials, for these uses of venetoclax with either hypomethylating agents or LDAC. The FDA also recently approved venetoclax in combination with rituximab (Rituxan) for treatment of patients who have chronic lymphocytic leukemia or small lymphocytic lymphoma, with or without 17p depletion, and have been treated with at least one prior therapy.
“AML is an aggressive disease with the lowest survival rate of all leukemias, and we look forward to working closely with the FDA to bring this potential option to patients with this very difficult-to-treat blood cancer as soon as possible,” said Sandra Horning, MD, chief medical officer at Genentech.
More information is included in the full release.
A supplemental new drug application (sNDA) for venetoclax (Venclexta) used in combination with either a hypomethylating agent or low-dose cytarabine (LDAC) for previously untreated acute myeloid leukemia has been submitted to the Food and Drug Administration by Genentech, which developed it.
Specifically, the sNDA is for these drug combinations in the treatment of AML patients ineligible for intensive chemotherapy, according to the announcement from Genentech.
The sNDA is based on results of two trials that included patients in this population. In the phase 1b M14-358 (NCT02203773), venetoclax was combined with either azacitidine or decitabine; patients treated with 400 mg of venetoclax had a complete remission rate of 73%, and the median overall survival across all doses of venetoclax was 17.5 months. Low white blood cell count with fever, low white blood cell count, anemia, low platelet count, and decreased potassium levels were the most common grade 3/4 adverse events (occurring in 10% or more of patients). In the phase 1b/2 study M14-387 (NCT02287233), venetoclax was used in combination with LDAC; patients treated with a 600-mg dose of venetoclax showed a complete response rate of 62%, and a median overall survival of 11.4 months. Low white blood cell count with fever, decreased potassium levels, pneumonia, disease progression, decreased phosphate levels, high blood pressure, and sepsis were the most common grade 3/4 adverse events seen in this study.
This sNDA follows FDA breakthrough therapy designations, based on these same trials, for these uses of venetoclax with either hypomethylating agents or LDAC. The FDA also recently approved venetoclax in combination with rituximab (Rituxan) for treatment of patients who have chronic lymphocytic leukemia or small lymphocytic lymphoma, with or without 17p depletion, and have been treated with at least one prior therapy.
“AML is an aggressive disease with the lowest survival rate of all leukemias, and we look forward to working closely with the FDA to bring this potential option to patients with this very difficult-to-treat blood cancer as soon as possible,” said Sandra Horning, MD, chief medical officer at Genentech.
More information is included in the full release.
A supplemental new drug application (sNDA) for venetoclax (Venclexta) used in combination with either a hypomethylating agent or low-dose cytarabine (LDAC) for previously untreated acute myeloid leukemia has been submitted to the Food and Drug Administration by Genentech, which developed it.
Specifically, the sNDA is for these drug combinations in the treatment of AML patients ineligible for intensive chemotherapy, according to the announcement from Genentech.
The sNDA is based on results of two trials that included patients in this population. In the phase 1b M14-358 (NCT02203773), venetoclax was combined with either azacitidine or decitabine; patients treated with 400 mg of venetoclax had a complete remission rate of 73%, and the median overall survival across all doses of venetoclax was 17.5 months. Low white blood cell count with fever, low white blood cell count, anemia, low platelet count, and decreased potassium levels were the most common grade 3/4 adverse events (occurring in 10% or more of patients). In the phase 1b/2 study M14-387 (NCT02287233), venetoclax was used in combination with LDAC; patients treated with a 600-mg dose of venetoclax showed a complete response rate of 62%, and a median overall survival of 11.4 months. Low white blood cell count with fever, decreased potassium levels, pneumonia, disease progression, decreased phosphate levels, high blood pressure, and sepsis were the most common grade 3/4 adverse events seen in this study.
This sNDA follows FDA breakthrough therapy designations, based on these same trials, for these uses of venetoclax with either hypomethylating agents or LDAC. The FDA also recently approved venetoclax in combination with rituximab (Rituxan) for treatment of patients who have chronic lymphocytic leukemia or small lymphocytic lymphoma, with or without 17p depletion, and have been treated with at least one prior therapy.
“AML is an aggressive disease with the lowest survival rate of all leukemias, and we look forward to working closely with the FDA to bring this potential option to patients with this very difficult-to-treat blood cancer as soon as possible,” said Sandra Horning, MD, chief medical officer at Genentech.
More information is included in the full release.
Protein could be target for MLL-rearranged AML
Preclinical research has revealed a potential therapeutic target for MLL-rearranged acute myeloid leukemia (AML).
The target—F-box protein S-phase kinase-associated protein 2 (Skp2)—degrades another protein called p27Kip1 that is important to the formation of healthy blood cells.
This finding was published in the Journal of Experimental Medicine.
“Our work provides a complete mechanistic look into the function of genetic and molecular programs driving this leukemia, and it exploits these processes to identify actionable therapeutic targets,’’ said study author H. Leighton Grimes, PhD, of Cincinnati Children’s Hospital Medical Center in Ohio.
For this work, Dr Grimes and his colleagues performed biochemical analyses of cells from AML patients. This gave the researchers comprehensive information about the targets and functions of the miR-196 molecular signaling pathway.
The team inserted mimics of miR-196 into MLL-AF9 leukemia cells to incorporate them into the cellular machinery. The group then lysed the cells for analyses, which revealed molecular targets of miR-196 in the leukemia cells.
Next, the researchers screened AML cells in mice for miR-196 targets. These experiments showed that certain microRNA targets are more important than others in the maintenance and spread of leukemia stem cells (LSCs).
Computer-assisted analysis of the Molecular Signature Database (a shared multi-institutional resource) allowed the researchers to identify sets of genes that show up in high numbers in MLL-AF9 leukemia.
Additional biochemical testing revealed that miR-196 directly targets and inhibits Cdkn1b/p27Kip1, which controls molecular programming in LSCs that allows them to maintain aggressive MLL-AF9 leukemia.
When miR-196 targets Cdkn1b/p27Kip1, it accelerates MLL-AF9 progression by abnormally linking stem cell activity with the growth of leukemia cells.
With the data suggesting that elevation of p27Kip1 protein levels may be therapeutic to AML patients, the researchers investigated a related molecular pathway that also regulates p27Kip1.
This investigation yielded the treatment target Skp2, which degrades the p27 protein and lowers its expression.
The researchers tested an experimental Skp2 inhibitor, SLZ P1041, on human AML cell lines and found the drug killed AML cells in a dose-dependent manner.
The team also tested SLZ P1041 in combination with other inhibitors—IBET-151, palbociclib, and MI-1. The most consistent synergies were with the combination of SLZ P1041 and MI-1, an inhibitor of the interaction between Menin and MLL.
“We still have extensive additional testing to conduct in laboratory animal models of AML before knowing if [targeting Skp2] will translate to patient care,” Dr Grimes said.
Preclinical research has revealed a potential therapeutic target for MLL-rearranged acute myeloid leukemia (AML).
The target—F-box protein S-phase kinase-associated protein 2 (Skp2)—degrades another protein called p27Kip1 that is important to the formation of healthy blood cells.
This finding was published in the Journal of Experimental Medicine.
“Our work provides a complete mechanistic look into the function of genetic and molecular programs driving this leukemia, and it exploits these processes to identify actionable therapeutic targets,’’ said study author H. Leighton Grimes, PhD, of Cincinnati Children’s Hospital Medical Center in Ohio.
For this work, Dr Grimes and his colleagues performed biochemical analyses of cells from AML patients. This gave the researchers comprehensive information about the targets and functions of the miR-196 molecular signaling pathway.
The team inserted mimics of miR-196 into MLL-AF9 leukemia cells to incorporate them into the cellular machinery. The group then lysed the cells for analyses, which revealed molecular targets of miR-196 in the leukemia cells.
Next, the researchers screened AML cells in mice for miR-196 targets. These experiments showed that certain microRNA targets are more important than others in the maintenance and spread of leukemia stem cells (LSCs).
Computer-assisted analysis of the Molecular Signature Database (a shared multi-institutional resource) allowed the researchers to identify sets of genes that show up in high numbers in MLL-AF9 leukemia.
Additional biochemical testing revealed that miR-196 directly targets and inhibits Cdkn1b/p27Kip1, which controls molecular programming in LSCs that allows them to maintain aggressive MLL-AF9 leukemia.
When miR-196 targets Cdkn1b/p27Kip1, it accelerates MLL-AF9 progression by abnormally linking stem cell activity with the growth of leukemia cells.
With the data suggesting that elevation of p27Kip1 protein levels may be therapeutic to AML patients, the researchers investigated a related molecular pathway that also regulates p27Kip1.
This investigation yielded the treatment target Skp2, which degrades the p27 protein and lowers its expression.
The researchers tested an experimental Skp2 inhibitor, SLZ P1041, on human AML cell lines and found the drug killed AML cells in a dose-dependent manner.
The team also tested SLZ P1041 in combination with other inhibitors—IBET-151, palbociclib, and MI-1. The most consistent synergies were with the combination of SLZ P1041 and MI-1, an inhibitor of the interaction between Menin and MLL.
“We still have extensive additional testing to conduct in laboratory animal models of AML before knowing if [targeting Skp2] will translate to patient care,” Dr Grimes said.
Preclinical research has revealed a potential therapeutic target for MLL-rearranged acute myeloid leukemia (AML).
The target—F-box protein S-phase kinase-associated protein 2 (Skp2)—degrades another protein called p27Kip1 that is important to the formation of healthy blood cells.
This finding was published in the Journal of Experimental Medicine.
“Our work provides a complete mechanistic look into the function of genetic and molecular programs driving this leukemia, and it exploits these processes to identify actionable therapeutic targets,’’ said study author H. Leighton Grimes, PhD, of Cincinnati Children’s Hospital Medical Center in Ohio.
For this work, Dr Grimes and his colleagues performed biochemical analyses of cells from AML patients. This gave the researchers comprehensive information about the targets and functions of the miR-196 molecular signaling pathway.
The team inserted mimics of miR-196 into MLL-AF9 leukemia cells to incorporate them into the cellular machinery. The group then lysed the cells for analyses, which revealed molecular targets of miR-196 in the leukemia cells.
Next, the researchers screened AML cells in mice for miR-196 targets. These experiments showed that certain microRNA targets are more important than others in the maintenance and spread of leukemia stem cells (LSCs).
Computer-assisted analysis of the Molecular Signature Database (a shared multi-institutional resource) allowed the researchers to identify sets of genes that show up in high numbers in MLL-AF9 leukemia.
Additional biochemical testing revealed that miR-196 directly targets and inhibits Cdkn1b/p27Kip1, which controls molecular programming in LSCs that allows them to maintain aggressive MLL-AF9 leukemia.
When miR-196 targets Cdkn1b/p27Kip1, it accelerates MLL-AF9 progression by abnormally linking stem cell activity with the growth of leukemia cells.
With the data suggesting that elevation of p27Kip1 protein levels may be therapeutic to AML patients, the researchers investigated a related molecular pathway that also regulates p27Kip1.
This investigation yielded the treatment target Skp2, which degrades the p27 protein and lowers its expression.
The researchers tested an experimental Skp2 inhibitor, SLZ P1041, on human AML cell lines and found the drug killed AML cells in a dose-dependent manner.
The team also tested SLZ P1041 in combination with other inhibitors—IBET-151, palbociclib, and MI-1. The most consistent synergies were with the combination of SLZ P1041 and MI-1, an inhibitor of the interaction between Menin and MLL.
“We still have extensive additional testing to conduct in laboratory animal models of AML before knowing if [targeting Skp2] will translate to patient care,” Dr Grimes said.
Identifying patients with a high risk of AML
Researchers say they have found ways to identify patients with a high risk of developing acute myeloid leukemia (AML).
The researchers used basic clinical and laboratory data to identify patients 6 to 12 months before AML presentation.
Using genetic information, the researchers were able to identify high-risk patients several years before AML presentation.
“This long time window gives us the first opportunity to think about how to prevent AML,” said John Dick, PhD, of Princess Margaret Cancer Centre in Toronto, Ontario, Canada.
However, Dr Dick and his colleagues noted that neither the genetic method nor the clinical data method were entirely accurate in identifying patients who would develop AML.
The researchers described both methods in Nature.
The team’s work began with the goal of distinguishing patients who will develop AML from patients who simply develop age-related clonal hematopoiesis.
“We wanted to know if there was any difference between these 2 groups in the genetics of their ‘normal’ blood samples taken at enrollment,” Dr Dick said. “To find out, we developed a gene-sequencing tool that captured the most common genes that get altered in AML and sequenced all the 500 blood samples.”
The researchers analyzed samples from 95 patients who ultimately developed AML and 414 age- and gender-matched controls. The samples from AML patients were obtained an average of 6.3 years before AML diagnosis.
The researchers found that pre-AML cases had more mutations per sample, higher variant allele frequencies, and a higher frequency of certain mutations than controls. Specifically, pre-AML cases had a significantly greater frequency of DNMT3A, TET2, SRSF2, ASXL1, TP53, U2AF1, JAK2, RUNX1, and IDH2 mutations.
The researchers used these findings to develop a model for predicting AML. They tested the model in a validation cohort of 29 pre-AML cases and 262 controls, as well as in a cohort combining the validation and discovery cohorts. The model predicted AML development with 41.9% sensitivity and 95.7% specificity.
However, the researchers said widespread use of this model would not be practical because AML is so rare. The team said millions of people would need to undergo screening to identify a few pre-AML cases, and there would be “many” false-positives.
Therefore, the researchers developed a model using clinical information from electronic health records—particularly blood counts. This model was able to predict AML development 6 to 12 months before diagnosis with a sensitivity of 25.7% and specificity of 98.2%.
The researchers said these results suggest clinical data can be used to identify patients with a high risk of AML who may benefit from targeted genetic screening. And combining clinical and genetic information in a single model could improve predictive accuracy.
“Our study provides, for the first time, evidence that we can identify people at risk of developing AML many years before they actually develop this life-threatening disease,” said study author George Vassiliou, PhD, of the Wellcome Trust Sanger Institute in Hinxton, UK.
“We hope to build on these findings to develop robust screening tests for identifying those at risk and drive research into how to prevent or stall progression towards AML. Our aspiration is that, one day, AML prevention would provide a compelling alternative to treatment.”
Researchers say they have found ways to identify patients with a high risk of developing acute myeloid leukemia (AML).
The researchers used basic clinical and laboratory data to identify patients 6 to 12 months before AML presentation.
Using genetic information, the researchers were able to identify high-risk patients several years before AML presentation.
“This long time window gives us the first opportunity to think about how to prevent AML,” said John Dick, PhD, of Princess Margaret Cancer Centre in Toronto, Ontario, Canada.
However, Dr Dick and his colleagues noted that neither the genetic method nor the clinical data method were entirely accurate in identifying patients who would develop AML.
The researchers described both methods in Nature.
The team’s work began with the goal of distinguishing patients who will develop AML from patients who simply develop age-related clonal hematopoiesis.
“We wanted to know if there was any difference between these 2 groups in the genetics of their ‘normal’ blood samples taken at enrollment,” Dr Dick said. “To find out, we developed a gene-sequencing tool that captured the most common genes that get altered in AML and sequenced all the 500 blood samples.”
The researchers analyzed samples from 95 patients who ultimately developed AML and 414 age- and gender-matched controls. The samples from AML patients were obtained an average of 6.3 years before AML diagnosis.
The researchers found that pre-AML cases had more mutations per sample, higher variant allele frequencies, and a higher frequency of certain mutations than controls. Specifically, pre-AML cases had a significantly greater frequency of DNMT3A, TET2, SRSF2, ASXL1, TP53, U2AF1, JAK2, RUNX1, and IDH2 mutations.
The researchers used these findings to develop a model for predicting AML. They tested the model in a validation cohort of 29 pre-AML cases and 262 controls, as well as in a cohort combining the validation and discovery cohorts. The model predicted AML development with 41.9% sensitivity and 95.7% specificity.
However, the researchers said widespread use of this model would not be practical because AML is so rare. The team said millions of people would need to undergo screening to identify a few pre-AML cases, and there would be “many” false-positives.
Therefore, the researchers developed a model using clinical information from electronic health records—particularly blood counts. This model was able to predict AML development 6 to 12 months before diagnosis with a sensitivity of 25.7% and specificity of 98.2%.
The researchers said these results suggest clinical data can be used to identify patients with a high risk of AML who may benefit from targeted genetic screening. And combining clinical and genetic information in a single model could improve predictive accuracy.
“Our study provides, for the first time, evidence that we can identify people at risk of developing AML many years before they actually develop this life-threatening disease,” said study author George Vassiliou, PhD, of the Wellcome Trust Sanger Institute in Hinxton, UK.
“We hope to build on these findings to develop robust screening tests for identifying those at risk and drive research into how to prevent or stall progression towards AML. Our aspiration is that, one day, AML prevention would provide a compelling alternative to treatment.”
Researchers say they have found ways to identify patients with a high risk of developing acute myeloid leukemia (AML).
The researchers used basic clinical and laboratory data to identify patients 6 to 12 months before AML presentation.
Using genetic information, the researchers were able to identify high-risk patients several years before AML presentation.
“This long time window gives us the first opportunity to think about how to prevent AML,” said John Dick, PhD, of Princess Margaret Cancer Centre in Toronto, Ontario, Canada.
However, Dr Dick and his colleagues noted that neither the genetic method nor the clinical data method were entirely accurate in identifying patients who would develop AML.
The researchers described both methods in Nature.
The team’s work began with the goal of distinguishing patients who will develop AML from patients who simply develop age-related clonal hematopoiesis.
“We wanted to know if there was any difference between these 2 groups in the genetics of their ‘normal’ blood samples taken at enrollment,” Dr Dick said. “To find out, we developed a gene-sequencing tool that captured the most common genes that get altered in AML and sequenced all the 500 blood samples.”
The researchers analyzed samples from 95 patients who ultimately developed AML and 414 age- and gender-matched controls. The samples from AML patients were obtained an average of 6.3 years before AML diagnosis.
The researchers found that pre-AML cases had more mutations per sample, higher variant allele frequencies, and a higher frequency of certain mutations than controls. Specifically, pre-AML cases had a significantly greater frequency of DNMT3A, TET2, SRSF2, ASXL1, TP53, U2AF1, JAK2, RUNX1, and IDH2 mutations.
The researchers used these findings to develop a model for predicting AML. They tested the model in a validation cohort of 29 pre-AML cases and 262 controls, as well as in a cohort combining the validation and discovery cohorts. The model predicted AML development with 41.9% sensitivity and 95.7% specificity.
However, the researchers said widespread use of this model would not be practical because AML is so rare. The team said millions of people would need to undergo screening to identify a few pre-AML cases, and there would be “many” false-positives.
Therefore, the researchers developed a model using clinical information from electronic health records—particularly blood counts. This model was able to predict AML development 6 to 12 months before diagnosis with a sensitivity of 25.7% and specificity of 98.2%.
The researchers said these results suggest clinical data can be used to identify patients with a high risk of AML who may benefit from targeted genetic screening. And combining clinical and genetic information in a single model could improve predictive accuracy.
“Our study provides, for the first time, evidence that we can identify people at risk of developing AML many years before they actually develop this life-threatening disease,” said study author George Vassiliou, PhD, of the Wellcome Trust Sanger Institute in Hinxton, UK.
“We hope to build on these findings to develop robust screening tests for identifying those at risk and drive research into how to prevent or stall progression towards AML. Our aspiration is that, one day, AML prevention would provide a compelling alternative to treatment.”
Oral arsenic/ATRA is noninferior to IV form in APML
For patients with non–high-risk acute promyelocytic leukemia (APML), the combination of an oral arsenic formulation and all-trans retinoic acid (ATRA) was noninferior to standard therapy with intravenous arsenic trioxide and ATRA, results of a randomized phase 3 trial show.
Among 109 patients with APML from one of 14 centers in China, the 2-year event-free survival rate after a median follow-up of 32 months was 97% for patients randomized to receive oral arsenic realgar-Indigo naturalis formula (RIF) plus ATRA, and 94% for patients randomized to IV arsenic trioxide plus ATRA, reported Hong-Hu Zhu, MD, of Peking University People’s Hospital in Beijing, China, and his colleagues.
“Our results suggest that non–high-risk acute promyelocytic leukemia can be cured using complete oral arsenic plus ATRA without conventional chemotherapy,” the investigators wrote. The report was published in The Lancet Oncology. “Although longer-term follow-up is needed to draw firm conclusions, our results support previously reported clinical and experimental evidence indicating that ATRA and arsenic act synergistically to eradicate acute promyelocytic leukemia.”
The combination of IV arsenic trioxide and ATRA has revolutionized the care of patients with APML, producing complete and durable remissions in more than 95% of patients with non–high-risk disease, defined as white blood cell counts of 10 x 109/L or less. The trial was designed to see whether an easier-to-administer all-oral regimen could be similarly efficacious and safe, the investigators said.
A total of 109 patients with newly diagnosed APML were randomly assigned on 2:1 basis to receive either RIF-ATRA (72 patients) or arsenic trioxide ATRA (37). Three patients in the oral arm and one in the arsenic trioxide arm did not receive the assigned therapy, but instead received ATRA and chemotherapy.
For induction, RIF was delivered 60 mg/kg daily in an oral divided dose; arsenic trioxide was delivered 0.15 mg/kg daily in an IV. ATRA was delivered 25 mg/m2 daily in an oral divided dose. Treatments were used until complete remissions were achieved.
Consolidation was home based and consisted of the same daily doses of RIF or arsenic trioxide in a 4-week-on/4-week-off regimen for four cycles, plus ATRA in the same daily dose in a 2-week-on/2-week-off regimen for seven cycles.
In a modified intention-to-treat analysis with 105 patients, 2-year EFS rates (the primary endpoint) were 97% with oral arsenic and 94% with arsenic trioxide. The percentage difference in EFS was 2.7% and RIF met the prespecified requirement for noninferiority because the lower limit of the 95% confidence interval (-5.8%) was greater than the noninferiority margin of –10%. The noninferiority of the oral formulation was confirmed in a per-protocol analysis, the investigators noted.
Grade 3 or 4 hepatotoxicities during induction were seen in 9% of patients treated with RIF-ATRA versus 14% of patients in the arsenic trioxide–ATRA group. Grade 3 or 4 infections occurred in 23% and 42% of patients, respectively.
Two patients in the arsenic trioxide–ATRA group died during induction therapy, one from hemorrhage and one from thrombocytopenia. There were no deaths during induction in the RIF-ATRA arm and no additional deaths in either arm during the consolidation phase.
All of the 103 surviving patients achieved complete remissions after consolidation.
The investigators acknowledged that the study was limited by a median follow-up time that was too short to allow definitive conclusions about overall survival. They plan to compare the costs of the two regimens in a future study.
SOURCE: Zhu HH et al. Lancet Oncol 2018;19:871-9.
The study by Dr. Zhu and colleagues advances the field of acute promyelocytic leukaemia treatment, but it also exposes important challenges in safely monitoring patients. Experienced hematologic oncologists are aware of the significant potential toxicity from arsenic treatment, including differentiation syndrome and thrombohemorrhagic events. Due to these potential complications, patients should always be managed in hospital during the first 2-3 weeks of induction, at least, so that they have the benefit of clinical and laboratory monitoring, transfusion support, and rapid supportive and therapeutic measures.
Francesco Lo-Coco, MD, and Laura Cicconi, MD, are with University Tor Vergata in Rome. Dr. Lo-Coco reported consultancy and speaker honoraria from Teva and Orsenix. Dr. Cicconi reported speaker honoraria from Teva. Their remarks are adapted and condensed from an accompanying editorial (Lancet Oncol. 2018;19:846-7).
The study by Dr. Zhu and colleagues advances the field of acute promyelocytic leukaemia treatment, but it also exposes important challenges in safely monitoring patients. Experienced hematologic oncologists are aware of the significant potential toxicity from arsenic treatment, including differentiation syndrome and thrombohemorrhagic events. Due to these potential complications, patients should always be managed in hospital during the first 2-3 weeks of induction, at least, so that they have the benefit of clinical and laboratory monitoring, transfusion support, and rapid supportive and therapeutic measures.
Francesco Lo-Coco, MD, and Laura Cicconi, MD, are with University Tor Vergata in Rome. Dr. Lo-Coco reported consultancy and speaker honoraria from Teva and Orsenix. Dr. Cicconi reported speaker honoraria from Teva. Their remarks are adapted and condensed from an accompanying editorial (Lancet Oncol. 2018;19:846-7).
The study by Dr. Zhu and colleagues advances the field of acute promyelocytic leukaemia treatment, but it also exposes important challenges in safely monitoring patients. Experienced hematologic oncologists are aware of the significant potential toxicity from arsenic treatment, including differentiation syndrome and thrombohemorrhagic events. Due to these potential complications, patients should always be managed in hospital during the first 2-3 weeks of induction, at least, so that they have the benefit of clinical and laboratory monitoring, transfusion support, and rapid supportive and therapeutic measures.
Francesco Lo-Coco, MD, and Laura Cicconi, MD, are with University Tor Vergata in Rome. Dr. Lo-Coco reported consultancy and speaker honoraria from Teva and Orsenix. Dr. Cicconi reported speaker honoraria from Teva. Their remarks are adapted and condensed from an accompanying editorial (Lancet Oncol. 2018;19:846-7).
For patients with non–high-risk acute promyelocytic leukemia (APML), the combination of an oral arsenic formulation and all-trans retinoic acid (ATRA) was noninferior to standard therapy with intravenous arsenic trioxide and ATRA, results of a randomized phase 3 trial show.
Among 109 patients with APML from one of 14 centers in China, the 2-year event-free survival rate after a median follow-up of 32 months was 97% for patients randomized to receive oral arsenic realgar-Indigo naturalis formula (RIF) plus ATRA, and 94% for patients randomized to IV arsenic trioxide plus ATRA, reported Hong-Hu Zhu, MD, of Peking University People’s Hospital in Beijing, China, and his colleagues.
“Our results suggest that non–high-risk acute promyelocytic leukemia can be cured using complete oral arsenic plus ATRA without conventional chemotherapy,” the investigators wrote. The report was published in The Lancet Oncology. “Although longer-term follow-up is needed to draw firm conclusions, our results support previously reported clinical and experimental evidence indicating that ATRA and arsenic act synergistically to eradicate acute promyelocytic leukemia.”
The combination of IV arsenic trioxide and ATRA has revolutionized the care of patients with APML, producing complete and durable remissions in more than 95% of patients with non–high-risk disease, defined as white blood cell counts of 10 x 109/L or less. The trial was designed to see whether an easier-to-administer all-oral regimen could be similarly efficacious and safe, the investigators said.
A total of 109 patients with newly diagnosed APML were randomly assigned on 2:1 basis to receive either RIF-ATRA (72 patients) or arsenic trioxide ATRA (37). Three patients in the oral arm and one in the arsenic trioxide arm did not receive the assigned therapy, but instead received ATRA and chemotherapy.
For induction, RIF was delivered 60 mg/kg daily in an oral divided dose; arsenic trioxide was delivered 0.15 mg/kg daily in an IV. ATRA was delivered 25 mg/m2 daily in an oral divided dose. Treatments were used until complete remissions were achieved.
Consolidation was home based and consisted of the same daily doses of RIF or arsenic trioxide in a 4-week-on/4-week-off regimen for four cycles, plus ATRA in the same daily dose in a 2-week-on/2-week-off regimen for seven cycles.
In a modified intention-to-treat analysis with 105 patients, 2-year EFS rates (the primary endpoint) were 97% with oral arsenic and 94% with arsenic trioxide. The percentage difference in EFS was 2.7% and RIF met the prespecified requirement for noninferiority because the lower limit of the 95% confidence interval (-5.8%) was greater than the noninferiority margin of –10%. The noninferiority of the oral formulation was confirmed in a per-protocol analysis, the investigators noted.
Grade 3 or 4 hepatotoxicities during induction were seen in 9% of patients treated with RIF-ATRA versus 14% of patients in the arsenic trioxide–ATRA group. Grade 3 or 4 infections occurred in 23% and 42% of patients, respectively.
Two patients in the arsenic trioxide–ATRA group died during induction therapy, one from hemorrhage and one from thrombocytopenia. There were no deaths during induction in the RIF-ATRA arm and no additional deaths in either arm during the consolidation phase.
All of the 103 surviving patients achieved complete remissions after consolidation.
The investigators acknowledged that the study was limited by a median follow-up time that was too short to allow definitive conclusions about overall survival. They plan to compare the costs of the two regimens in a future study.
SOURCE: Zhu HH et al. Lancet Oncol 2018;19:871-9.
For patients with non–high-risk acute promyelocytic leukemia (APML), the combination of an oral arsenic formulation and all-trans retinoic acid (ATRA) was noninferior to standard therapy with intravenous arsenic trioxide and ATRA, results of a randomized phase 3 trial show.
Among 109 patients with APML from one of 14 centers in China, the 2-year event-free survival rate after a median follow-up of 32 months was 97% for patients randomized to receive oral arsenic realgar-Indigo naturalis formula (RIF) plus ATRA, and 94% for patients randomized to IV arsenic trioxide plus ATRA, reported Hong-Hu Zhu, MD, of Peking University People’s Hospital in Beijing, China, and his colleagues.
“Our results suggest that non–high-risk acute promyelocytic leukemia can be cured using complete oral arsenic plus ATRA without conventional chemotherapy,” the investigators wrote. The report was published in The Lancet Oncology. “Although longer-term follow-up is needed to draw firm conclusions, our results support previously reported clinical and experimental evidence indicating that ATRA and arsenic act synergistically to eradicate acute promyelocytic leukemia.”
The combination of IV arsenic trioxide and ATRA has revolutionized the care of patients with APML, producing complete and durable remissions in more than 95% of patients with non–high-risk disease, defined as white blood cell counts of 10 x 109/L or less. The trial was designed to see whether an easier-to-administer all-oral regimen could be similarly efficacious and safe, the investigators said.
A total of 109 patients with newly diagnosed APML were randomly assigned on 2:1 basis to receive either RIF-ATRA (72 patients) or arsenic trioxide ATRA (37). Three patients in the oral arm and one in the arsenic trioxide arm did not receive the assigned therapy, but instead received ATRA and chemotherapy.
For induction, RIF was delivered 60 mg/kg daily in an oral divided dose; arsenic trioxide was delivered 0.15 mg/kg daily in an IV. ATRA was delivered 25 mg/m2 daily in an oral divided dose. Treatments were used until complete remissions were achieved.
Consolidation was home based and consisted of the same daily doses of RIF or arsenic trioxide in a 4-week-on/4-week-off regimen for four cycles, plus ATRA in the same daily dose in a 2-week-on/2-week-off regimen for seven cycles.
In a modified intention-to-treat analysis with 105 patients, 2-year EFS rates (the primary endpoint) were 97% with oral arsenic and 94% with arsenic trioxide. The percentage difference in EFS was 2.7% and RIF met the prespecified requirement for noninferiority because the lower limit of the 95% confidence interval (-5.8%) was greater than the noninferiority margin of –10%. The noninferiority of the oral formulation was confirmed in a per-protocol analysis, the investigators noted.
Grade 3 or 4 hepatotoxicities during induction were seen in 9% of patients treated with RIF-ATRA versus 14% of patients in the arsenic trioxide–ATRA group. Grade 3 or 4 infections occurred in 23% and 42% of patients, respectively.
Two patients in the arsenic trioxide–ATRA group died during induction therapy, one from hemorrhage and one from thrombocytopenia. There were no deaths during induction in the RIF-ATRA arm and no additional deaths in either arm during the consolidation phase.
All of the 103 surviving patients achieved complete remissions after consolidation.
The investigators acknowledged that the study was limited by a median follow-up time that was too short to allow definitive conclusions about overall survival. They plan to compare the costs of the two regimens in a future study.
SOURCE: Zhu HH et al. Lancet Oncol 2018;19:871-9.
FROM THE LANCET ONCOLOGY
Key clinical point:
Major finding: The 2-year event-free survival was 97% with oral arsenic/ATRA versus 94% with IV arsenic trioxide/ATRA.
Study details: Randomized open-label phase 3 trial of 109 patients with APML.
Disclosures: The National Natural Science Foundation of China, the Beijing Municipal Science and Technology Commission, and the National Key R&D Program of China supported the study. The investigators reported having no financial disclosures.
Source: Zhu HH et al. Lancet Oncol. 2018;19:871-9.