A phase 1 trial of vadastuximab talirine combined with hypomethylating agents in patients with CD33-positive AML.

Treatment of acute myeloid leukemia (AML) among the elderly is challenging because of intolerance of intensive therapy and therapy-resistant biology. Hypomethylating agents (HMAs) are commonly used, with suboptimal outcomes. Vadastuximab talirine is a CD33-directed antibody conjugated to pyrrolobenzodiazepine (PBD) dimers. Preclinically, HMAs followed by vadastuximab talirine produced upregulated CD33 expression, increased DNA incorporation by PBD, and enhanced cytotoxicity. A combination cohort in a phase 1 study (NCT01902329) assessed safety, tolerability, and activity of vadastuximab talirine with HMAs. Those eligible had Eastern Cooperative Oncology Group status 0 to 1 and previously untreated CD33-positive AML, and declined intensive therapy. Vadastuximab talirine was administered intravenously at 10 μg/kg on last day of HMA (azacitidine or decitabine) infusion in 4-week cycles. Among 53 patients treated, the median age was 75 years. Patients had adverse (38%) or intermediate (62%) cytogenetic risk. Median treatment duration was 19.3 weeks. No dose-limiting toxicities were reported. The majority of adverse events were a result of myelosuppression, with some causing therapy delays. Thirty- and 60-day mortality rates were 2% and 8%, respectively. The composite remission rate (complete remission [CR] and CR with incomplete blood count recovery) was 70%. Fifty-one percent of remissions were minimal residual disease-negative by flow cytometry. Similarly high remission rates were observed in patients with secondary AML, aged at least 75 years, and with adverse cytogenetic risk. Median relapse-free survival and overall survival were 7.7 and 11.3 months, respectively. Compared with historical data for HMA monotherapy, the combination of vadastuximab talirine with HMAs produced a high remission rate, but was accompanied by increased hematologic toxicity.

[1]  D. Faller,et al.  Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, combined with azacitidine in patients with AML. , 2018, Blood.

[2]  H. Kantarjian,et al.  Phase Ib Study of Glasdegib, a Hedgehog Pathway Inhibitor, in Combination with Standard Chemotherapy in Patients with AML or High-Risk MDS , 2018, Clinical Cancer Research.

[3]  A. Letai,et al.  Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study. , 2018, The Lancet. Oncology.

[4]  F. Ravandi,et al.  A phase 1 trial of vadastuximab talirine as monotherapy in patients with CD33-positive acute myeloid leukemia. , 2018, Blood.

[5]  B. Goh,et al.  Phase 1 dose escalation multicenter trial of pracinostat alone and in combination with azacitidine in patients with advanced hematologic malignancies , 2017, Cancer.

[6]  P. Vyas,et al.  Mutant Isocitrate Dehydrogenase (mIDH) Inhibitors, Enasidenib or Ivosidenib, in Combination with Azacitidine (AZA): Preliminary Results of a Phase 1b/2 Study in Patients with Newly Diagnosed Acute Myeloid Leukemia (AML) , 2017 .

[7]  E. Estey,et al.  Allogeneic Hematopoietic Cell Transplantation for Acute Myeloid Leukemia: Time to Move Toward a Minimal Residual Disease-Based Definition of Complete Remission? , 2016, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[8]  J. Cortes,et al.  Final results of a phase III randomized trial of CPX-351 versus 7+3 in older patients with newly diagnosed high risk (secondary) AML. , 2016 .

[9]  M. Konopleva,et al.  A phase II study of decitabine and gemtuzumab ozogamicin in newly diagnosed and relapsed acute myeloid leukemia and high-risk myelodysplastic syndrome , 2016, Leukemia.

[10]  D. Benjamin,et al.  SGN-CD33A in Combination with Hypomethylating Agents Is Highly Efficacious in Preclinical Models of AML , 2015 .

[11]  R. Greil,et al.  International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. , 2015, Blood.

[12]  H. Lazarus,et al.  A phase I study of midostaurin and azacitidine in relapsed and elderly AML patients. , 2015, Clinical lymphoma, myeloma & leukemia.

[13]  H. Erba Finding the optimal combination therapy for the treatment of newly diagnosed AML in older patients unfit for intensive therapy. , 2015, Leukemia research.

[14]  G. Ossenkoppele,et al.  How I treat the older patient with acute myeloid leukemia. , 2015, Blood.

[15]  J. Paulson,et al.  Siglec-mediated regulation of immune cell function in disease , 2014, Nature Reviews Immunology.

[16]  E. Estey,et al.  Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. , 2014, The Lancet. Oncology.

[17]  D. Howard,et al.  A phase 2 trial of azacitidine and gemtuzumab ozogamicin therapy in older patients with acute myeloid leukemia. , 2013, Blood.

[18]  R. Larson,et al.  A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. , 2013, Blood.

[19]  Jorge Cortes,et al.  Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. , 2013, Blood.

[20]  B. Wood,et al.  Randomized, phase IIb study of low-dose cytarabine and lintuzumab versus low-dose cytarabine and placebo in older adults with untreated acute myeloid leukemia , 2013, Haematologica.

[21]  M. Minden,et al.  Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[22]  R. Hills,et al.  Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. , 2010, Blood.

[23]  B. Wood,et al.  Complete remissions observed in acute myeloid leukemia following prolonged exposure to lintuzumab: a phase 1 trial , 2009, Leukemia & lymphoma.

[24]  C. Bennett,et al.  Gemtuzumab ozogamicin-associated sinusoidal obstructive syndrome (SOS): an overview from the research on adverse drug events and reports (RADAR) project. , 2007, Leukemia research.

[25]  I. Bernstein,et al.  Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33‐positive acute myeloid leukemia in first recurrence , 2005, Cancer.

[26]  D. Scheinberg,et al.  Phase III randomized multicenter study of a humanized anti-CD33 monoclonal antibody, lintuzumab, in combination with chemotherapy, versus chemotherapy alone in patients with refractory or first-relapsed acute myeloid leukemia. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[27]  C. Bloomfield,et al.  Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. , 2003, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  A. Gewirtz,et al.  Gemtuzumab ozogamicin (Mylotarg) monotherapy for relapsed AML after hematopoietic stem cell transplant: efficacy and incidence of hepatic veno-occlusive disease , 2002, Bone Marrow Transplantation.

[29]  G. McDonald,et al.  Hepatic sinusoidal obstruction after gemtuzumab ozogamicin (Mylotarg) therapy. , 2002, Blood.

[30]  I. Bernstein,et al.  Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. , 2001, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[31]  Daniel J Weisdorf,et al.  Acute Myeloid Leukemia. , 2015, The New England journal of medicine.

[32]  R. Hills,et al.  The addition of gemtuzumab ozogamicin to low-dose Ara-C improves remission rate but does not significantly prolong survival in older patients with acute myeloid leukaemia: results from the LRF AML14 and NCRI AML16 pick-a-winner comparison , 2013, Leukemia.