A first-in-human phase I trial of NKG2D chimeric antigen receptor-T cells in AML/MDS and multiple myeloma.

Authors

null

Sarah Nikiforow

Dana-Farber Cancer Institute, Boston, MA

Sarah Nikiforow , Joana Murad , Heather Daley , Helene Negre , Jake Reder , Charles L. Sentman , Frederic Lehmann , Sarah Snykers , Rachel Allen , Ilene Galinsky , Nikhil C. Munshi , Richard M. Stone , Robert Soiffer , Jerome Ritz , Susanne H.C. Baumeister

Organizations

Dana-Farber Cancer Institute, Boston, MA, Celdara Medical, LLC, Lebanon, NH, Geisel School of Medicine at Dartmouth, Lebanon, NH, Celyad, SA, Mont Saint Guibert, Belgium, Dana-Farber Cancer Instititue/Children's Hosp Boston, Boston, MA

Research Funding

Pharmaceutical/Biotech Company

Background: Canonical CAR-T cell constructs encode a single chain antibody variable fragment, costimulatory domain, and signaling domain of CD3 zeta. This restricts recognition to 1 tumor antigen and a limited set of cancers. This study employs a novel CAR fusing full-length human Natural Killer Group 2D (NKG2D) gene with the human CD3 zeta signaling domain. NKG2D CAR receives costimulation via naturally-expressed DAP10 and activates T cells directly to kill and secrete cytokines upon recognition of MIC-A, MIC-B, and UL-16 binding proteins. These NKG2D-ligands are upregulated in many solid tumors and hematologic malignancies but absent or poorly expressed on healthy tissues. In multiple murine cancer models, NKG2D CAR-T cells induced complete remissions, T-cell memory, and altered tumor microenvironment via cytokines. We demonstrated manufacture of autologous NKG2D CAR-T cells in a GMP-environment from healthy adults and patients with AML and myeloma was feasible. Following isolation of mononuclear cells, T cells are activated with anti-CD3 mAb and IL-2, undergo 2 transductions with SFG retroviral vector containing NKG2D CAR construct, and expand in media containing IL-2. Validation studies yielded consistent viability, robust cell expansion, high vector-driven NKG2D expression on T cells, potent IFN-g production during tumor cell co-culture, viral copy number/cell < 5, and no replication-competent retrovirus in NKG2D CAR-T (CM-CS1) cells. Methods: A phase 1 dose-escalation study to establish safety and feasibility of a single infusion of CM-CS1 T cells without lymphodepleting conditioning is currently enrolling subjects with AML/MDS-RAEB or relapsed/refractory progressive multiple myeloma without standard therapy options (ClinicalTrials.gov NCT02203825). Dose-escalation will proceed in 4 cohorts [1x106 to 3x107 CM-CS1 T cells] according to a 3+3 design followed by expansion cohorts at the MTDs in AML/MDS and myeloma for an anticipated 24 subjects. As of February, 2016, 6 subjects have been treated. Cohort 1 was completed without DLTs. Additional studies may include lymphodepletion, multiple infusions, and cytoreductive chemotherapy in both hematologic and solid tumor malignancies. Clinical trial information: NCT02203825

Disclaimer

This material on this page is ©2024 American Society of Clinical Oncology, all rights reserved. Licensing available upon request. For more information, please contact licensing@asco.org

Abstract Details

Meeting

2016 ASCO Annual Meeting

Session Type

Poster Session

Session Title

Developmental Therapeutics—Immunotherapy

Track

Developmental Therapeutics and Translational Research

Sub Track

Cellular Immunotherapy

Clinical Trial Registration Number

NCT02203825

Citation

J Clin Oncol 34, 2016 (suppl; abstr TPS3102)

DOI

10.1200/JCO.2016.34.15_suppl.TPS3102

Abstract #

TPS3102

Poster Bd #

420b

Abstract Disclosures

Similar Abstracts

First Author: Fuchun Guo

First Author: Yan Luo

Abstract

2020 ASCO Virtual Scientific Program

Eradication of medulloblastoma by NKG2D-specific CAR T-cells.

First Author: Hong-jiu Dai