A Phase I/II Study of Evofosfamide, A Hypoxia-activated Prodrug with or without Bortezomib in Subjects with Relapsed/Refractory Multiple Myeloma

Purpose: The presence of hypoxia in the diseased bone marrow presents a new therapeutic target for multiple myeloma. Evofosfamide (formerly TH-302) is a 2-nitroimidazole prodrug of the DNA alkylator, bromo-isophosphoramide mustard, which is selectively activated under hypoxia. This trial was designed as a phase I/II study investigating evofosfamide in combination with dexamethasone, and in combination with bortezomib and dexamethasone in relapsed/refractory multiple myeloma. Patients and Methods: Fifty-nine patients initiated therapy, 31 received the combination of evofosfamide and dexamethasone, and 28 received the combination of evofosfamide, bortezomib, and dexamethasone. Patients were heavily pretreated with a median number of prior therapies of 7 (range: 2–15). All had previously received bortezomib and immunomodulators. The MTD, treatment toxicity, and efficacy were determined. Results: The MTD was established at 340 mg/m2 evofosfamide + dexamethasone with dose-limiting mucositis at higher doses. For the combination of evofosfamide, bortezomib, and dexamethasone, no patient had a dose-limiting toxicity (DLT) and the recommended phase II dose was established at 340 mg/m2. The most common ≥grade 3 adverse events (AE) were thrombocytopenia (25 patients), anemia (24 patients), neutropenia (15 patients), and leukopenia (9 patients). Skin toxicity was reported in 42 (71%) patients. Responses included 1 very good partial response (VGPR), 3 partial response (PR), 2 minor response (MR), 20 stable disease (SD), and 4 progressive disease (PD) for evofosfamide + dexamethasone and 1 complete response (CR), 2 PR, 1 MR, 18 SD, and 5 PD for evofosfamide + bortezomib + dexamethasone. Disease stabilization was observed in over 80% and this was reflective of the prolonged overall survival of 11.2 months. Conclusions: Evofosfamide can be administered at 340 mg/m2 twice a week with or without bortezomib. Clinical activity has been noted in patients with heavily pretreated relapsed refractory multiple myeloma.

[1]  D. Steensma,et al.  The bone-marrow niche in MDS and MGUS: implications for AML and MM , 2018, Nature Reviews Clinical Oncology.

[2]  K. Anderson,et al.  Management of relapsed and refractory multiple myeloma: novel agents, antibodies, immunotherapies and beyond , 2017, Leukemia.

[3]  D. Landau,et al.  Genomic complexity of multiple myeloma and its clinical implications , 2017, Nature Reviews Clinical Oncology.

[4]  H. Goldschmidt,et al.  Daratumumab, Lenalidomide, and Dexamethasone for Multiple Myeloma. , 2016, The New England journal of medicine.

[5]  H. Goldschmidt,et al.  International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. , 2016, The Lancet. Oncology.

[6]  A. Palumbo,et al.  Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. , 2016, The New England journal of medicine.

[7]  F. Prósper,et al.  Targeting vasculogenesis to prevent progression in multiple myeloma , 2016, Leukemia.

[8]  R. Lewensohn,et al.  First-in-human, phase I/IIa clinical study of the peptidase potentiated alkylator melflufen administered every three weeks to patients with advanced solid tumor malignancies , 2015, Investigational New Drugs.

[9]  Jessica Katz,et al.  Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. , 2015, The New England journal of medicine.

[10]  M. Borad,et al.  Randomized Phase II Trial of Gemcitabine Plus TH-302 Versus Gemcitabine in Patients With Advanced Pancreatic Cancer. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[11]  Simion I. Chiosea,et al.  Genomic Correlate of Exceptional Erlotinib Response in Head and Neck Squamous Cell Carcinoma. , 2015, JAMA oncology.

[12]  G. Görgün,et al.  Targeting the bone marrow microenvironment in multiple myeloma , 2015, Immunological reviews.

[13]  Hans Erik Johnsen,et al.  International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. , 2014, The Lancet. Oncology.

[14]  K. Ganjoo,et al.  Phase II study of the safety and antitumor activity of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[15]  A. Azab,et al.  Hypoxia Promotes Dissemination and Colonization in New Bone Marrow Niches in Waldenström Macroglobulinemia , 2014, Molecular Cancer Research.

[16]  A. Roccaro,et al.  Role of endothelial progenitor cells in cancer progression. , 2014, Biochimica et biophysica acta.

[17]  D. Quail,et al.  Microenvironmental regulation of tumor progression and metastasis , 2014 .

[18]  J. Massagué,et al.  Hypoxia signaling--license to metastasize. , 2013, Cancer discovery.

[19]  C. Hart,et al.  Synergistic Induction of Apoptosis in Multiple Myeloma Cells by Bortezomib and Hypoxia-Activated Prodrug TH-302, In Vivo and In Vitro , 2013, Molecular Cancer Therapeutics.

[20]  P. L. Bergsagel,et al.  Drug response in a genetically engineered mouse model of multiple myeloma is predictive of clinical efficacy. , 2012, Blood.

[21]  H. Goldschmidt,et al.  Risk of progression and survival in multiple myeloma relapsing after therapy with IMiDs and bortezomib: A multicenter international myeloma working group study , 2012, Leukemia.

[22]  Damien J. Ferraro,et al.  TH-302, a hypoxia-activated prodrug with broad in vivo preclinical combination therapy efficacy: optimization of dosing regimens and schedules , 2012, Cancer Chemotherapy and Pharmacology.

[23]  Damien J. Ferraro,et al.  Selective Tumor Hypoxia Targeting by Hypoxia-Activated Prodrug TH-302 Inhibits Tumor Growth in Preclinical Models of Cancer , 2011, Clinical Cancer Research.

[24]  P. Glazer,et al.  Molecular and Cellular Pharmacology of the Hypoxia-Activated Prodrug TH-302 , 2011, Molecular Cancer Therapeutics.

[25]  Charles P. Lin,et al.  Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. , 2011, Blood.

[26]  G. Roodman,et al.  Combination of bendamustine, lenalidomide, and dexamethasone (BLD) in patients with relapsed or refractory multiple myeloma is feasible and highly effective: results of phase 1/2 open-label, dose escalation study. , 2011, Blood.

[27]  G. Semenza,et al.  Hypoxia-inducible factor 1 is a master regulator of breast cancer metastatic niche formation , 2011, Proceedings of the National Academy of Sciences.

[28]  Fei Xing,et al.  Hypoxia-induced Jagged2 promotes breast cancer metastasis and self-renewal of cancer stem-like cells , 2011, Oncogene.

[29]  Suzanne F. Jones,et al.  Phase 1 Study of the Safety, Tolerability, and Pharmacokinetics of TH-302, a Hypoxia-Activated Prodrug, in Patients with Advanced Solid Malignancies , 2011, Clinical Cancer Research.

[30]  D. Hanahan,et al.  Hallmarks of Cancer: The Next Generation , 2011, Cell.

[31]  Z. Duan,et al.  Role of hypoxia and glycolysis in the development of multi-drug resistance in human tumor cells and the establishment of an orthotopic multi-drug resistant tumor model in nude mice using hypoxic pre-conditioning , 2011, Cancer Cell International.

[32]  D. Ribatti,et al.  Low bone marrow oxygen tension and hypoxia-inducible factor-1α overexpression characterize patients with multiple myeloma: role on the transcriptional and proangiogenic profiles of CD138+ cells , 2010, Leukemia.

[33]  C. Hart,et al.  Targeting the multiple myeloma hypoxic niche with TH-302, a hypoxia-activated prodrug. , 2010, Blood.

[34]  G. Tonon,et al.  Targeting angiogenesis via a c-Myc/hypoxia-inducible factor-1alpha-dependent pathway in multiple myeloma. , 2009, Cancer research.

[35]  R. Hoffman,et al.  Potent and highly selective hypoxia-activated achiral phosphoramidate mustards as anticancer drugs. , 2008, Journal of medicinal chemistry.

[36]  Kenneth C. Anderson,et al.  Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets , 2007, Nature Reviews Cancer.

[37]  B. Barlogie,et al.  International uniform response criteria for multiple myeloma , 2006, Leukemia.

[38]  C. Hofmeister,et al.  NCCN Guidelines® Insights: Multiple Myeloma, Version 3.2022. , 2022, Journal of the National Comprehensive Cancer Network : JNCCN.

[39]  D. Dingli,et al.  Safety Outcomes for Autologous Stem Cell Transplant in Multiple Myeloma , 2018, Mayo Clinic proceedings.

[40]  C. Hofmeister,et al.  NCCN Guidelines Insights: Multiple Myeloma, Version 3.2018. , 2018, Journal of the National Comprehensive Cancer Network : JNCCN.