Final results of a phase 2 clinical trial of LCL161, an oral SMAC mimetic for patients with myelofibrosis.

Outcomes in patients with high-risk and treatment-resistant myelofibrosis (MF) post-JAK inhibitor therapy remain poor, with no approved drug therapies beyond the JAK inhibitor class. In certain clinical situations, such as severe thrombocytopenia, administration of most JAK inhibitors are contraindicated. Thus, there is an unmet medical need for the development of novel agents for patients with MF. SMAC mimetics [or inhibitor of apoptosis (IAP) antagonists] induce apoptosis in cancer cells. Because these agents are hypothesized to have increased activity in a tumor necrosis factor-α cytokine-rich microenvironment, as is the case with MF, we conducted a single-center, investigator-initiated phase 2 clinical trial, with a monovalent SMAC mimetic LCL161 (oral, starting dose, 1500 mg per week) in patients with intermediate to high-risk MF. In an older group, 66% with ≥2 prior therapies and a median baseline platelet count of 52 × 103/μL and 28% with ASXL1 mutations, we observed a 30% objective response by Revised International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) 2013 criteria. Notably, 6 responding patients achieved clinical improvement of anemia: 4, hemoglobin response; 2, transfusion independence. Median OS was 34 months (range, 2.2-60.1+). Reductions of cIAPs were observed in all responders. The most common toxicity was nausea/vomiting (N/V) in 64% (mostly grade 1/2); fatigue in 46%; and dizziness/vertigo in 30%. There were 4 grade 3/4 adverse events (2, syncope; 1, N/V; 1, skin eruption/pruritis). There were 2 deaths during the study period, both unrelated to the study drug. SMAC mimetics may represent an option for older patients with thrombocytopenia or for those in whom prior JAK inhibitors has failed. This trial was registered at www.clinicaltrials.gov as #NCT02098161.

[1]  S. Verstovsek,et al.  Management of myelofibrosis after ruxolitinib failure , 2020, Leukemia & lymphoma.

[2]  R. Mesa,et al.  Management of myelofibrosis after ruxolitinib failure , 2020, Annals of Hematology.

[3]  Angela G. Fleischman,et al.  The SMAC mimetic LCL-161 selectively targets JAK2V617F mutant cells , 2020, Experimental Hematology & Oncology.

[4]  R. Foà,et al.  Life after ruxolitinib: Reasons for discontinuation, impact of disease phase, and outcomes in 218 patients with myelofibrosis , 2019, Cancer.

[5]  T. Aittokallio,et al.  Integrated drug profiling and CRISPR screening identify essential pathways for CAR T cell cytotoxicity. , 2019, Blood.

[6]  H. Kestler,et al.  Biomarker profile for prediction of response to SMAC mimetic monotherapy in pediatric precursor B‐cell acute lymphoblastic leukemia , 2019, International journal of cancer.

[7]  S. Verstovsek,et al.  Novel Therapies in Myeloproliferative Neoplasms (MPN): Beyond JAK Inhibitors , 2019, Current Hematologic Malignancy Reports.

[8]  A. Rademaker,et al.  Aurora Kinase A Inhibition Provides Clinical Benefit, Normalizes Megakaryocytes, and Reduces Bone Marrow Fibrosis in Patients with Myelofibrosis: A Phase I Trial , 2019, Clinical Cancer Research.

[9]  S. Verstovsek,et al.  SMAC mimetics as potential cancer therapeutics in myeloid malignancies , 2019, British journal of haematology.

[10]  P. Fenaux,et al.  Luspatercept for the treatment of anemia in myelodysplastic syndromes and primary myelofibrosis. , 2019, Blood.

[11]  E. Engle,et al.  Cytokine production in myelofibrosis exhibits differential responsiveness to JAK-STAT, MAP kinase, and NFκB signaling , 2019, Leukemia.

[12]  S. Verstovsek,et al.  New Concepts of Treatment for Patients with Myelofibrosis , 2019, Current Treatment Options in Oncology.

[13]  F. Hernandez-Ilizaliturri,et al.  The SMAC mimetic LCL-161 displays antitumor activity in preclinical models of rituximab-resistant B-cell lymphoma. , 2018, Blood advances.

[14]  Sung-Bae Kim,et al.  Paclitaxel With Inhibitor of Apoptosis Antagonist, LCL161, for Localized Triple-Negative Breast Cancer, Prospectively Stratified by Gene Signature in a Biomarker-Driven Neoadjuvant Trial. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[15]  R. Bell,et al.  Autocrine Tnf signaling favors malignant cells in myelofibrosis in a Tnfr2-dependent fashion , 2018, Leukemia.

[16]  J. Lancet,et al.  Between a rux and a hard place: evaluating salvage treatment and outcomes in myelofibrosis after ruxolitinib discontinuation , 2018, Annals of Hematology.

[17]  M. Cazzola,et al.  MIPSS70: Mutation-Enhanced International Prognostic Score System for Transplantation-Age Patients With Primary Myelofibrosis. , 2017, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[18]  H. Kantarjian,et al.  Clonal evolution and outcomes in myelofibrosis after ruxolitinib discontinuation. , 2017, Blood.

[19]  Z. Shun,et al.  Janus kinase-2 inhibitor fedratinib in patients with myelofibrosis previously treated with ruxolitinib (JAKARTA-2): a single-arm, open-label, non-randomised, phase 2, multicentre study. , 2017, The Lancet. Haematology.

[20]  T. Alain,et al.  Smac mimetics synergize with immune checkpoint inhibitors to promote tumour immunity against glioblastoma , 2017, Nature Communications.

[21]  Y. Asmann,et al.  IAP antagonists induce anti-tumor immunity in multiple myeloma , 2016, Nature Medicine.

[22]  Shaji K. Kumar,et al.  Smac mimetic LCL161 overcomes protective ER stress induced by obatoclax, synergistically causing cell death in multiple myeloma , 2016, Oncotarget.

[23]  J. Cortes,et al.  Safety and Efficacy of Fedratinib in Patients With Primary or Secondary Myelofibrosis: A Randomized Clinical Trial. , 2015, JAMA oncology.

[24]  J. Infante,et al.  Phase I dose-escalation study of LCL161, an oral inhibitor of apoptosis proteins inhibitor, in patients with advanced solid tumors. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[25]  S. Fulda Targeting inhibitor of apoptosis proteins for cancer therapy: a double-edge sword? , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[26]  S. Fulda,et al.  Inhibitor of Apoptosis (IAP) proteins in hematological malignancies: molecular mechanisms and therapeutic opportunities , 2014, Leukemia.

[27]  J. Mascarenhas Rationale for combination therapy in myelofibrosis. , 2014, Best practice & research. Clinical haematology.

[28]  Yigong Shi,et al.  Birinapant (TL32711), a Bivalent SMAC Mimetic, Targets TRAF2-Associated cIAPs, Abrogates TNF-Induced NF-κB Activation, and Is Active in Patient-Derived Xenograft Models , 2014, Molecular Cancer Therapeutics.

[29]  James M. Bogenberger,et al.  Synergistic targeting of AML stem/progenitor cells with IAP antagonist birinapant and demethylating agents. , 2014, Journal of the National Cancer Institute.

[30]  G. Häcker,et al.  Inhibitors of apoptosis proteins (IAPs) are required for effective T-cell expansion/survival during antiviral immunity in mice. , 2014, Blood.

[31]  N. Kröger,et al.  Revised response criteria for myelofibrosis: International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European LeukemiaNet (ELN) consensus report. , 2013, Blood.

[32]  M. Andreeff,et al.  Safety and pharmacokinetics of the antisense oligonucleotide (ASO) LY2181308 as a single-agent or in combination with idarubicin and cytarabine in patients with refractory or relapsed acute myeloid leukemia (AML) , 2013, Investigational New Drugs.

[33]  Jason Gotlib,et al.  A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. , 2012, The New England journal of medicine.

[34]  T. Barbui,et al.  JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. , 2012, The New England journal of medicine.

[35]  Simone Fulda,et al.  Targeting IAP proteins for therapeutic intervention in cancer , 2012, Nature Reviews Drug Discovery.

[36]  Angela G. Fleischman,et al.  TNFα facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. , 2011, Blood.

[37]  T. Barbui,et al.  The Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF): international prospective validation and reliability trial in 402 patients. , 2011, Blood.

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

[39]  F. Passamonti,et al.  DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[40]  N. Hacohen,et al.  IAP inhibitors enhance co-stimulation to promote tumor immunity , 2010, The Journal of experimental medicine.

[41]  D. Porter,et al.  Smac mimetics: implications for enhancement of targeted therapies in leukemia , 2010, Leukemia.

[42]  Z. Estrov,et al.  Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. , 2010, The New England journal of medicine.

[43]  Stephen J. Morris,et al.  Phase I/II trial of AEG35156 X-linked inhibitor of apoptosis protein antisense oligonucleotide combined with idarubicin and cytarabine in patients with relapsed or primary refractory acute myeloid leukemia. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[44]  Xiaodong Wang,et al.  Smac, a Mitochondrial Protein that Promotes Cytochrome c–Dependent Caspase Activation by Eliminating IAP Inhibition , 2000, Cell.

[45]  P. Thall,et al.  Bayesian sequential monitoring designs for single-arm clinical trials with multiple outcomes. , 1995, Statistics in medicine.