1
|
Dam S, Tscherne A, Engels L, Sutter G, Osterhaus ADME, Rimmelzwaan GF. Design and evaluation of a poly-epitope based vaccine for the induction of influenza A virus cross-reactive CD8 + T cell responses. Sci Rep 2025; 15:10586. [PMID: 40148547 PMCID: PMC11950192 DOI: 10.1038/s41598-025-95479-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
The availability of influenza vaccines that can induce broadly protective immune responses is highly desirable and could also mitigate the impact of future influenza pandemics. Ideally, these vaccines also induce virus-specific CD8 + T cells, which have been identified as an independent correlate of protection. In the present study, we explored the use of an artificial immunogen that comprises of twenty highly conserved influenza virus CD8 + T cell epitopes with an HLA coverage of 99.5% of the world population. The highly attenuated viral vector Modified Vaccinia virus Ankara (MVA) was used to deliver the artificial poly-epitope sequence (rMVA-PE) and by using T cell lines raised against individual epitopes, we confirmed that the epitopes are liberated from the artificial immunogen. For efficient antigen processing and presentation, the epitopes were separated by spacer sequences. Stimulation of peripheral blood mononuclear cells of HLA-typed blood donors with rMVA-PE resulted in the activation of influenza virus-specific T cell responses. Furthermore, immunization of humanized HLA-A2.1-/HLA-DR1-transgenic H-2 class I-/class II-knockout mice (HLA-A*02:01) with rMVA-PE induced influenza virus-specific CD8 + T cell responses. Thus, rMVA-PE proved to be immunogenic both in vitro and in vivo and constitutes a promising vaccine candidate for the induction of cross-reactive CD8 + T cell responses that could afford protection against antigenically distinct influenza A viruses (IAV) of various subtypes and species, and is currently considered for further clinical testing.
Collapse
Affiliation(s)
- Sharmistha Dam
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Alina Tscherne
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), Oberschleißheim, Germany
- German Center for Infection Research (DZIF), partner site Munich, Oberschleißheim, Germany
| | - Leoni Engels
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University Munich (LMU Munich), Oberschleißheim, Germany
- German Center for Infection Research (DZIF), partner site Munich, Oberschleißheim, Germany
| | - Albert D M E Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany.
| |
Collapse
|
2
|
Sun L, Romancik JT. The Development and Application of Bispecific Antibodies in B-Cell Non-Hodgkin Lymphoma. J Pers Med 2025; 15:51. [PMID: 39997328 PMCID: PMC11856678 DOI: 10.3390/jpm15020051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/08/2025] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
T-cell-engaging bispecific antibodies (BsAbs) are monoclonal antibodies that redirect the cytotoxic activity of T-cells to target malignant neoplasms. B-cell non-Hodgkin lymphoma (B-NHL) is a heterogenous group of aggressive and indolent malignancies with significant therapeutic challenges due to high relapse rates and limited options for relapsed/refractory disease. BsAbs function by simultaneously binding to CD3 on endogenous T-cells and a tumor-associated antigen, creating an immunologic synapse which results in the death of the target cell. The widespread T-cell activation that occurs with BsAb administration can result in cytokine release syndrome and neurological adverse events. Mosunetuzumab, epcoritamab, and glofitamab are CD20-targeting BsAbs that have demonstrated promising single-agent activity in both indolent and aggressive B-NHL. BsAbs are now being evaluated in combination with other anti-lymphoma agents and in earlier lines of treatment, and the results of ongoing clinical trials involving these agents have the potential to reshape the treatment landscape for B-NHL. In this review, we describe the structural features, clinical data, and toxicity profile associated with the BsAbs currently used to treat B-NHL and then discuss ongoing studies and future directions for this exciting new class of therapeutic agents.
Collapse
Affiliation(s)
- Laura Sun
- Department of Pharmacy, Winship Cancer Institute of Emory University, Atlanta, GA 30308, USA;
| | - Jason T. Romancik
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30308, USA
| |
Collapse
|
3
|
Mog BJ, Marcou N, DiNapoli SR, Pearlman AH, Nichakawade TD, Hwang MS, Douglass J, Hsiue EHC, Glavaris S, Wright KM, Konig MF, Paul S, Wyhs N, Ge J, Miller MS, Azurmendi P, Watson E, Pardoll DM, Gabelli SB, Bettegowda C, Papadopoulos N, Kinzler KW, Vogelstein B, Zhou S. Preclinical studies show that Co-STARs combine the advantages of chimeric antigen and T cell receptors for the treatment of tumors with low antigen densities. Sci Transl Med 2024; 16:eadg7123. [PMID: 38985855 DOI: 10.1126/scitranslmed.adg7123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/01/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
Two types of engineered T cells have been successfully used to treat patients with cancer, one with an antigen recognition domain derived from antibodies [chimeric antigen receptors (CARs)] and the other derived from T cell receptors (TCRs). CARs use high-affinity antigen-binding domains and costimulatory domains to induce T cell activation but can only react against target cells with relatively high amounts of antigen. TCRs have a much lower affinity for their antigens but can react against target cells displaying only a few antigen molecules. Here, we describe a new type of receptor, called a Co-STAR (for costimulatory synthetic TCR and antigen receptor), that combines aspects of both CARs and TCRs. In Co-STARs, the antigen-recognizing components of TCRs are replaced by high-affinity antibody fragments, and costimulation is provided by two modules that drive NF-κB signaling (MyD88 and CD40). Using a TCR-mimic antibody fragment that targets a recurrent p53 neoantigen presented in a common human leukocyte antigen (HLA) allele, we demonstrate that T cells equipped with Co-STARs can kill cancer cells bearing low densities of antigen better than T cells engineered with conventional CARs and patient-derived TCRs in vitro. In mouse models, we show that Co-STARs mediate more robust T cell expansion and more durable tumor regressions than TCRs similarly modified with MyD88 and CD40 costimulation. Our data suggest that Co-STARs may have utility for other peptide-HLA antigens in cancer and other targets where antigen density may limit the efficacy of engineered T cells.
Collapse
Affiliation(s)
- Brian J Mog
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Nikita Marcou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah R DiNapoli
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alexander H Pearlman
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tushar D Nichakawade
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | - Michael S Hwang
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jacqueline Douglass
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Emily Han-Chung Hsiue
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Stephanie Glavaris
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Katharine M Wright
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Maximilian F Konig
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Suman Paul
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Nicolas Wyhs
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jiaxin Ge
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michelle S Miller
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - P Azurmendi
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Evangeline Watson
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Sandra B Gabelli
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chetan Bettegowda
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nickolas Papadopoulos
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kenneth W Kinzler
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Bert Vogelstein
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| |
Collapse
|
4
|
Zhou X, Geyer FK, Happel D, Takimoto J, Kolmar H, Rabinovich B. Using protein geometry to optimize cytotoxicity and the cytokine window of a ROR1 specific T cell engager. Front Immunol 2024; 15:1323049. [PMID: 38455046 PMCID: PMC10917902 DOI: 10.3389/fimmu.2024.1323049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
T cell engaging bispecific antibodies have shown clinical proof of concept for hematologic malignancies. Still, cytokine release syndrome, neurotoxicity, and on-target-off-tumor toxicity, especially in the solid tumor setting, represent major obstacles. Second generation TCEs have been described that decouple cytotoxicity from cytokine release by reducing the apparent binding affinity for CD3 and/or the TAA but the results of such engineering have generally led only to reduced maximum induction of cytokine release and often at the expense of maximum cytotoxicity. Using ROR1 as our model TAA and highly modular camelid nanobodies, we describe the engineering of a next generation decoupled TCE that incorporates a "cytokine window" defined as a dose range in which maximal killing is reached but cytokine release may be modulated from very low for safety to nearly that induced by first generation TCEs. This latter attribute supports pro-inflammatory anti-tumor activity including bystander killing and can potentially be used by clinicians to safely titrate patient dose to that which mediates maximum efficacy that is postulated as greater than that possible using standard second generation approaches. We used a combined method of optimizing TCE mediated synaptic distance and apparent affinity tuning of the TAA binding arms to generate a relatively long but persistent synapse that supports a wide cytokine window, potent killing and a reduced propensity towards immune exhaustion. Importantly, this next generation TCE induced significant tumor growth inhibition in vivo but unlike a first-generation non-decoupled benchmark TCE that induced lethal CRS, no signs of adverse events were observed.
Collapse
Affiliation(s)
- Xueyuan Zhou
- Drug Discovery and Development, Fuse Biotherapeutics, Woburn, MA, United States
| | - Felix Klaus Geyer
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Dominic Happel
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Jeffrey Takimoto
- Drug Discovery and Development, Fuse Biotherapeutics, Woburn, MA, United States
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Brian Rabinovich
- Drug Discovery and Development, Fuse Biotherapeutics, Woburn, MA, United States
| |
Collapse
|
5
|
Robertson IB, Mulvaney R, Dieckmann N, Vantellini A, Canestraro M, Amicarella F, O'Dwyer R, Cole DK, Harper S, Dushek O, Kirk P. Tuning the potency and selectivity of ImmTAC molecules by affinity modulation. Clin Exp Immunol 2024; 215:105-119. [PMID: 37930865 PMCID: PMC10847821 DOI: 10.1093/cei/uxad120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/08/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
T-cell-engaging bispecifics have great clinical potential for the treatment of cancer and infectious diseases. The binding affinity and kinetics of a bispecific molecule for both target and T-cell CD3 have substantial effects on potency and specificity, but the rules governing these relationships are not fully understood. Using immune mobilizing monoclonal TCRs against cancer (ImmTAC) molecules as a model, we explored the impact of altering affinity for target and CD3 on the potency and specificity of the redirected T-cell response. This class of bispecifics binds specific target peptides presented by human leukocyte antigen on the cell surface via an affinity-enhanced T-cell receptor and can redirect T-cell activation with an anti-CD3 effector moiety. The data reveal that combining a strong affinity TCR with an intermediate affinity anti-CD3 results in optimal T-cell activation, while strong affinity of both targeting and effector domains significantly reduces maximum cytokine release. Moreover, by optimizing the affinity of both parts of the molecule, it is possible to improve the selectivity. These results could be effectively modelled based on kinetic proofreading with limited signalling. This model explained the experimental observation that strong binding at both ends of the molecules leads to reduced activity, through very stable target-bispecific-effector complexes leading to CD3 entering a non-signalling dark state. These findings have important implications for the design of anti-CD3-based bispecifics with optimal biophysical parameters for both activity and specificity.
Collapse
Affiliation(s)
- Ian B Robertson
- Immunocore Limited, Drug Discovery and Protein Engineering, Abingdon, Oxon, UK
| | - Rachel Mulvaney
- Immunocore Limited, Drug Discovery and Protein Engineering, Abingdon, Oxon, UK
| | - Nele Dieckmann
- Immunocore Limited, Drug Discovery and Protein Engineering, Abingdon, Oxon, UK
| | - Alessio Vantellini
- Immunocore Limited, Drug Discovery and Protein Engineering, Abingdon, Oxon, UK
| | - Martina Canestraro
- Immunocore Limited, Drug Discovery and Protein Engineering, Abingdon, Oxon, UK
| | | | - Ronan O'Dwyer
- Immunocore Limited, Drug Discovery and Protein Engineering, Abingdon, Oxon, UK
| | - David K Cole
- Immunocore Limited, Drug Discovery and Protein Engineering, Abingdon, Oxon, UK
| | - Stephen Harper
- Immunocore Limited, Drug Discovery and Protein Engineering, Abingdon, Oxon, UK
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Peter Kirk
- Immunocore Limited, Drug Discovery and Protein Engineering, Abingdon, Oxon, UK
| |
Collapse
|
6
|
Mohammadian Gol T, Kim M, Sinn R, Ureña-Bailén G, Stegmeyer S, Gratz PG, Zahedipour F, Roig-Merino A, Antony JS, Mezger M. CRISPR-Cas9-Based Gene Knockout of Immune Checkpoints in Expanded NK Cells. Int J Mol Sci 2023; 24:16065. [PMID: 38003255 PMCID: PMC10671270 DOI: 10.3390/ijms242216065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Natural killer (NK) cell immunotherapy has emerged as a novel treatment modality for various cancer types, including leukemia. The modulation of inhibitory signaling pathways in T cells and NK cells has been the subject of extensive investigation in both preclinical and clinical settings in recent years. Nonetheless, further research is imperative to optimize antileukemic activities, especially regarding NK-cell-based immunotherapies. The central scientific question of this study pertains to the potential for boosting cytotoxicity in expanded and activated NK cells through the inhibition of inhibitory receptors. To address this question, we employed the CRISPR-Cas9 system to target three distinct inhibitory signaling pathways in NK cells. Specifically, we examined the roles of A2AR within the metabolic purinergic signaling pathway, CBLB as an intracellular regulator in NK cells, and the surface receptors NKG2A and CD96 in enhancing the antileukemic efficacy of NK cells. Following the successful expansion of NK cells, they were transfected with Cas9+sgRNA RNP to knockout A2AR, CBLB, NKG2A, and CD96. The analysis of indel frequencies for all four targets revealed good knockout efficiencies in expanded NK cells, resulting in diminished protein expression as confirmed by flow cytometry and Western blot analysis. Our in vitro killing assays demonstrated that NKG2A and CBLB knockout led to only a marginal improvement in the cytotoxicity of NK cells against AML and B-ALL cells. Furthermore, the antileukemic activity of CD96 knockout NK cells did not yield significant enhancements, and the blockade of A2AR did not result in significant improvement in killing efficiency. In conclusion, our findings suggest that CRISPR-Cas9-based knockout strategies for immune checkpoints might not be sufficient to efficiently boost the antileukemic functions of expanded (and activated) NK cells and, at the same time, point to the need for strong cellular activating signals, as this can be achieved, for example, via transgenic chimeric antigen receptor expression.
Collapse
Affiliation(s)
- Tahereh Mohammadian Gol
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Miso Kim
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Ralph Sinn
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Guillermo Ureña-Bailén
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Sarah Stegmeyer
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Paul Gerhard Gratz
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Fatemeh Zahedipour
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | | | - Justin S. Antony
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| | - Markus Mezger
- Department of General Pediatrics, Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany; (T.M.G.); (M.K.); (P.G.G.); (F.Z.)
| |
Collapse
|
7
|
Lee E, Lee S, Park S, Son YG, Yoo J, Koh Y, Shin DY, Lim Y, Won J. Asymmetric anti-CLL-1×CD3 bispecific antibody, ABL602 2+1, with attenuated CD3 affinity endows potent antitumor activity but limited cytokine release. J Immunother Cancer 2023; 11:e007494. [PMID: 37848261 PMCID: PMC10582864 DOI: 10.1136/jitc-2023-007494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a type of leukemia in adults with a high mortality rate and poor prognosis. Although targeted therapeutics, chemotherapy, and hematopoietic stem cell transplantation can improve the prognosis, the recurrence rate is still high, with a 5-year survival rate of approximately 40%. This study aimed to develop an IgG-based asymmetric bispecific antibody that targets CLL-1 and CD3 for treating AML. METHODS ABL602 candidates were compared in terms of binding activity, T-cell activation, and tumor-killing activities. ABL602-mediated T-cell activation and tumor-killing activities were determined by measuring the expression of activation markers, cytokines, cytolytic proteins, and the proportion of dead cells. We evaluated in vivo tumor growth inhibitory activity in two mouse models bearing subcutaneously and orthotopically engrafted human AML. Direct tumor-killing activity and T-cell activation in patient-derived AML blasts were also evaluated. RESULTS ABL602 2+1 showed a limited CD3 binding in the absence of CLL-1, suggesting that steric hindrance on the CD3 binding arm could reduce CLL-1 expression-independent CD3 binding. Although the CD3 binding activity was attenuated compared with that of 1+1, ABL602 2+1 exhibited much stronger T-cell activation and potent tumor-killing activities in AML cell lines. ABL602 2+1 efficiently inhibited tumor progression in subcutaneously and orthotopically engrafted AML mouse models. In the orthotopic mouse model, tumor growth inhibition was observed by gross measurement of luciferase activity, as well as a reduced proportion of AML blasts in the bone marrow, as determined by flow cytometry and immunohistochemistry (IHC) staining. ABL602 2+1 efficiently activated T cells and induced the lysis of AML blasts, even at very low effector:target (E:T) ratios (eg, 1:50). Compared with the reference 1+1 antibody, ABL602 did not induce the release of cytokines including interleukin-6 and tumor necrosis factor-α in the healthy donor-derived peripheral blood mononuclear cell. CONCLUSIONS With its potent tumor-killing activity and reduced cytokine release, ABL602 2+1 is a promising candidate for treating patients with AML and warrants further study.
Collapse
Affiliation(s)
- Eunhee Lee
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Shinai Lee
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| | | | | | - Jiseon Yoo
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Korea (the Republic of)
| | - Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Korea (the Republic of)
| | - Yangmi Lim
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Jonghwa Won
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| |
Collapse
|
8
|
Yun K, Siegler EL, Kenderian SS. Who wins the combat, CAR or TCR? Leukemia 2023; 37:1953-1962. [PMID: 37626090 DOI: 10.1038/s41375-023-01976-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/04/2023] [Accepted: 07/17/2023] [Indexed: 08/27/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has drawn increasing attention over the last few decades given its remarkable effectiveness and breakthroughs in treating B cell hematological malignancies. Even though CAR-T cell therapy has outstanding clinical successes, most treated patients still relapse after infusion. CARs are derived from the T cell receptor (TCR) complex and co-stimulatory molecules associated with T cell activation; however, the similarities and differences between CARs and endogenous TCRs regarding their sensitivity, signaling pathway, killing mechanisms, and performance are still not fully understood. In this review, we discuss the parallel comparisons between CARs and TCRs from various aspects and how these current findings might provide novel insights and contribute to improvement of CAR-T cell therapy efficacy.
Collapse
Affiliation(s)
- Kun Yun
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Nowill AE, Caruso M, de Campos-Lima PO. T-cell immunity to SARS-CoV-2: what if the known best is not the optimal course for the long run? Adapting to evolving targets. Front Immunol 2023; 14:1133225. [PMID: 37388738 PMCID: PMC10303130 DOI: 10.3389/fimmu.2023.1133225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/11/2023] [Indexed: 07/01/2023] Open
Abstract
Humanity did surprisingly well so far, considering how unprepared it was to respond to the coronavirus disease 2019 (COVID-19) threat. By blending old and ingenious new technology in the context of the accumulated knowledge on other human coronaviruses, several vaccine candidates were produced and tested in clinical trials in record time. Today, five vaccines account for the bulk of the more than 13 billion doses administered worldwide. The ability to elicit biding and neutralizing antibodies most often against the spike protein is a major component of the protection conferred by immunization but alone it is not enough to limit virus transmission. Thus, the surge in numbers of infected individuals by newer variants of concern (VOCs) was not accompanied by a proportional increase in severe disease and death rate. This is likely due to antiviral T-cell responses, whose evasion is more difficult to achieve. The present review helps navigating the very large literature on T cell immunity induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and vaccination. We examine the successes and shortcomings of the vaccinal protection in the light of the emergence of VOCs with breakthrough potential. SARS-CoV-2 and human beings will likely coexist for a long while: it will be necessary to update existing vaccines to improve T-cell responses and attain better protection against COVID-19.
Collapse
Affiliation(s)
- Alexandre E. Nowill
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas, SP, Brazil
| | - Manuel Caruso
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, Québec, QC, Canada
| | - Pedro O. de Campos-Lima
- Boldrini Children’s Center, Campinas, SP, Brazil
- Molecular and Morphofunctional Biology Graduate Program, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
10
|
Yang Y, Yang H, Alcaina Y, Puc J, Birt A, Vedvyas Y, Gallagher M, Alla S, Riascos MC, McCloskey JE, Du K, Gonzalez-Valdivieso J, Min IM, de Stanchina E, Britz M, von Hofe E, Jin MM. Inducible expression of interleukin-12 augments the efficacy of affinity-tuned chimeric antigen receptors in murine solid tumor models. Nat Commun 2023; 14:2068. [PMID: 37045815 PMCID: PMC10097865 DOI: 10.1038/s41467-023-37646-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
The limited number of targetable tumor-specific antigens and the immunosuppressive nature of the microenvironment within solid malignancies represent major barriers to the success of chimeric antigen receptor (CAR)-T cell therapies. Here, using epithelial cell adhesion molecule (EpCAM) as a model antigen, we used alanine scanning of the complementarity-determining region to fine-tune CAR affinity. This allowed us to identify CARs that could spare primary epithelial cells while still effectively targeting EpCAMhigh tumors. Although affinity-tuned CARs showed suboptimal antitumor activity in vivo, we found that inducible secretion of interleukin-12 (IL-12), under the control of the NFAT promoter, can restore CAR activity to levels close to that of the parental CAR. This strategy was further validated with another affinity-tuned CAR specific for intercellular adhesion molecule-1 (ICAM-1). Only in affinity-tuned CAR-T cells was NFAT activity stringently controlled and restricted to tumors expressing the antigen of interest at high levels. Our study demonstrates the feasibility of specifically gearing CAR-T cells towards recognition of solid tumors by combining inducible IL-12 expression and affinity-tuned CAR.
Collapse
Affiliation(s)
- Yanping Yang
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Huan Yang
- AffyImmune Therapeutics, Inc., Natick, MA, 01760, USA
| | - Yago Alcaina
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Janusz Puc
- AffyImmune Therapeutics, Inc., Natick, MA, 01760, USA
| | - Alyssa Birt
- AffyImmune Therapeutics, Inc., Natick, MA, 01760, USA
| | - Yogindra Vedvyas
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | | | - Srinija Alla
- AffyImmune Therapeutics, Inc., Natick, MA, 01760, USA
| | - Maria Cristina Riascos
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jaclyn E McCloskey
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Karrie Du
- AffyImmune Therapeutics, Inc., Natick, MA, 01760, USA
| | - Juan Gonzalez-Valdivieso
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Irene M Min
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Matt Britz
- AffyImmune Therapeutics, Inc., Natick, MA, 01760, USA
| | - Eric von Hofe
- AffyImmune Therapeutics, Inc., Natick, MA, 01760, USA
| | - Moonsoo M Jin
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA.
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
11
|
|
12
|
Kühl L, Schäfer AK, Kraft S, Aschmoneit N, Kontermann RE, Seifert O. eIg-based bispecific T-cell engagers targeting EGFR: Format matters. MAbs 2023; 15:2183540. [PMID: 36864566 PMCID: PMC9988351 DOI: 10.1080/19420862.2023.2183540] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Bispecific antibodies are molecules with versatile modes of action and applications for therapy. They are commonly developed as T-cell engagers (TCE), which simultaneously target an antigen expressed by tumor cells and CD3 expressed by T-cells, thereby inducing T-cell-mediated target cell killing. There is growing evidence that the molecular composition and valency for the target antigen influence the activity of TCEs. Here, the eIg platform technology was used to generate a set of bispecific TCEs targeting epidermal growth factor receptors (EGFR) and CD3. These molecules either included or lacked an Fc region and exhibited one binding site for CD3 and either one or two binding sites for EGFR (1 + 1 or 2 + 1 formats) utilizing different molecular arrangements of the binding sites. In total, 11 different TCE formats were analyzed for binding to target cells and T cells, T cell-mediated killing of tumor cells, and for the activation of T cells (release of cytokines and proliferation of T-cells). Bivalent binding to EGFR strongly increased binding and T cell-mediated killing. However, the molecular composition and position of the CD3-binding arm also affected target cell killing, cytokine release, and T-cell proliferation. Our findings support that screening of a panel of formats is beneficial to identify the most potent bispecific TCE, and that format matters.
Collapse
Affiliation(s)
- Lennart Kühl
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Annelie K Schäfer
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Sebastian Kraft
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Nadine Aschmoneit
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
13
|
Ball K, Dovedi SJ, Vajjah P, Phipps A. Strategies for clinical dose optimization of T cell-engaging therapies in oncology. MAbs 2023; 15:2181016. [PMID: 36823042 PMCID: PMC9980545 DOI: 10.1080/19420862.2023.2181016] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Innovative approaches in the design of T cell-engaging (TCE) molecules are ushering in a new wave of promising immunotherapies for the treatment of cancer. Their mechanism of action, which generates an in trans interaction to create a synthetic immune synapse, leads to complex and interconnected relationships between the exposure, efficacy, and toxicity of these drugs. Challenges thus arise when designing optimal clinical dose regimens for TCEs with narrow therapeutic windows, with a variety of dosing strategies being evaluated to mitigate key side effects such as cytokine release syndrome, neurotoxicity, and on-target off-tumor toxicities. This review evaluates the current approaches to dose optimization throughout the preclinical and clinical development of TCEs, along with perspectives for improvement of these strategies. Quantitative approaches used to aid the understanding of dose-exposure-response relationships are highlighted, along with opportunities to guide the rational design of next-generation TCE molecules, and optimize their dose regimens in patients.
Collapse
Affiliation(s)
- Kathryn Ball
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Pavan Vajjah
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Alex Phipps
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
14
|
Long M, Mims AS, Li Z. Factors Affecting the Cancer Immunotherapeutic Efficacy of T Cell Bispecific Antibodies and Strategies for Improvement. Immunol Invest 2022; 51:2176-2214. [PMID: 36259611 DOI: 10.1080/08820139.2022.2131569] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
T-cell bispecific antibodies (T-BsAbs) are a new class of cancer immunotherapy drugs that can simultaneously bind to tumor-associated antigens on target cells and to the CD3 subunit of the T-cell receptor (TCR) on T cells. In the last decade, numerous T-BsAbs have been developed for the treatment of both hematological malignancies and solid tumors. Among them, blinatumomab has been successfully used to treat CD19 positive malignancies and has been approved by the FDA as standard care for acute lymphoblastic leukemia (ALL). However, in many clinical scenarios, the efficacy of T-BsAbs remains unsatisfactory. To further improve T-BsAb therapy, it will be crucial to better understand the factors affecting treatment efficacy and the nature of the T-BsAb-induced immune response. Herein, we first review the studies on the potential mechanisms by which T-BsAbs activate T-cells and how they elicit efficient target killing despite suboptimal costimulatory support. We focus on analyzing reports from clinical trials and preclinical studies, and summarize the factors that have been identified to impact the efficacy of T-BsAbs. Lastly, we review current and propose new approaches to improve the clinical efficacy of T-BsAbs.
Collapse
Affiliation(s)
- Meixiao Long
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.,Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Alice S Mims
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.,Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|
15
|
Su X, Sun T, Li M, Xia Y, Li M, Wang D, Lu F, Ye J, Ji C. Lkb1 aggravates diffuse large B-cell lymphoma by promoting the function of Treg cells and immune escape. Lab Invest 2022; 20:378. [PMID: 35986288 PMCID: PMC9392310 DOI: 10.1186/s12967-022-03588-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/12/2022] [Indexed: 12/03/2022]
Abstract
Background Regulatory T cells (Tregs) induce immune responses and may contribute to immune escape in tumors. Accumulation of Tregs in tumors represents a critical barrier to anti-tumor immunity and immunotherapy. However, conflicting results describing the role of Tregs in lymphoma warrant further investigation. The precise features and mechanisms underlying the alteration in Tregs in diffuse large B-cell lymphoma (DLBCL) are not well understood yet. In this study, we analyzed the mechanism underlying the observed alterations in Tregs in DLBCL and examined the effect of Lkb1 expression on the immunosuppressive function of human Tregs. Methods Flow cytometry and immunofluorescence were used to analyze the proportion of Tregs and effector Tregs in the peripheral blood and lymph nodes of patients with DLBCL and control group. In vitro culture assays were used to analyze the immunosuppressive function of Tregs in the two groups. Transcriptome sequencing was performed to analyze the differentially expressed genes in the two groups, and the transcription level and protein expression of Lkb1 in the two groups were detected using RT-PCR and WES microprotein technology. Lentiviral vectors were constructed to explore the functional changes of Tregs with stable upregulation and downregulation of Lkb1. Finally, a humanized murine lymphoma model was established to study the function of Lkb1 in Tregs in the pathogenesis of DLBCL. Results The number of Tregs was found to be dramatically increased in peripheral blood and tumor tissue in DLBCL patients compared with that in healthy controls, and decreased after treatment. Tregs from DLBCL patients exhibited multiple enhanced functions, including increased inhibition of CD8+cytotoxic T cells (CTL) against tumor cells, enhanced suppression of CD8+CTL secretion of granular enzyme, and suppression of CD8+CTL degranulation. Lkb1 was found to be upregulated in Tregs of DLBCL patients. Furthermore, Lkb1 contributes to Treg immunosuppressive function in DLBCL by regulating the mevalonate pathway. Finally, deletion of Lkb1 in Tregs suppressed tumor growth and promoted anti-tumor immunity in a DLBCL murine model. Conclusions These findings confirmed that Lkb1-regulated Tregs are critical for immune escape in DLBCL, which emphasizes that Lkb1 is a potential target for the immunotherapy of DLBCL. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03588-0.
Collapse
|
16
|
McCue AC, Yao Z, Kuhlman B. Advances in modular control of CAR-T therapy with adapter-mediated CARs. Adv Drug Deliv Rev 2022; 187:114358. [PMID: 35618140 PMCID: PMC9939278 DOI: 10.1016/j.addr.2022.114358] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/11/2022] [Accepted: 05/18/2022] [Indexed: 11/01/2022]
Abstract
Protein engineering has contributed to successes in the field of T cell-based immunotherapy, including chimeric antigen receptor (CAR) T cell therapy. CAR T cell therapy has become a pillar of cancer immunotherapy, demonstrating clinical effectiveness against B cell malignancies by targeting the B cell antigen CD19. Current gene editing techniques have limited safety controls over CAR T cell activity, which presents a hurdle for control of CAR T cells in patients. Alternatively, CAR T cell activity can be controlled by engineering CARs to bind soluble adapter molecules that direct the interaction between the CAR T cell and target cell. The flexibility in this adapter-mediated approach overcomes the rigid specificity of traditional CAR T cells to allow targeting of multiple cell types. Here we describe adapter CAR T technologies and how these methods emphasize the growing role of protein engineering in the design of programmable tools for T cell therapies.
Collapse
Affiliation(s)
- Amelia C McCue
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Zhiyuan Yao
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27514, USA.
| |
Collapse
|
17
|
Sun Y, Dong Y, Sun R, Liu Y, Wang Y, Luo H, Shi B, Jiang H, Li Z. Chimeric anti-GPC3 sFv-CD3ε receptor-modified T cells with IL7 co-expression for the treatment of solid tumors. Mol Ther Oncolytics 2022; 25:160-173. [PMID: 35572194 PMCID: PMC9065615 DOI: 10.1016/j.omto.2022.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/14/2022] [Indexed: 11/20/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells targeting glypican-3 (GPC3) demonstrated early signs of therapeutic efficacy to hepatocellular carcinoma patients with a risk of cytokine release syndrome (CRS). Several adoptive cell therapies (ACTs) with T cells using the natural T cell receptor (TCR) signaling induced more efficient antitumor function and reduced cytokine production relative to CARs in solid tumors. To improve the efficacy and safety of GPC3-targeted ACTs, T cells were modified with anti-GPC3 single-chain fragment variable(sFv) linked to CD3ε, which could be incorporated into the entire TCR/CD3 complex to form chimeric sFv-CD3ε receptor (sFv-ε). sFv-ε T cells showed competitive antitumor activity and lower cytokine release compared to 28ζ or BBζ CAR T cells, which may be ascribed to moderately less activated Ca2+-calcineurin-NFAT signaling pathway. We further generated murine sFv-ε T cells with interleukin-7 co-expression (7sFv-ε) to promote T cell survival and to mobilize the endogenous immune system. In immunocompetent mouse models, 7sFv-ε T cells showed superior persistence, antitumor efficacy, and immunological memory while preserving the low production of cytokines associated with CRS compared to conventional sFv-ε T cells. These results indicate that GPC3-specific 7sFv-ε T cells could serve as a promising therapeutic strategy for solid tumors.
Collapse
Affiliation(s)
- Yansha Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Yiwei Dong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Ruixin Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Yifan Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Yi Wang
- CARsgen Therapeutics, Shanghai 200032, China
| | - Hong Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiaotong University, Shanghai 200032, China
| | - Bizhi Shi
- CARsgen Therapeutics, Shanghai 200032, China
| | - Hua Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China.,CARsgen Therapeutics, Shanghai 200032, China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China.,CARsgen Therapeutics, Shanghai 200032, China
| |
Collapse
|
18
|
McKenzie B, Khazen R, Valitutti S. Greek Fire, Poison Arrows, and Scorpion Bombs: How Tumor Cells Defend Against the Siege Weapons of Cytotoxic T Lymphocytes. Front Immunol 2022; 13:894306. [PMID: 35592329 PMCID: PMC9110820 DOI: 10.3389/fimmu.2022.894306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/07/2022] [Indexed: 01/05/2023] Open
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) are the main cellular effectors of the adaptive immune response against cancer cells, which in turn have evolved sophisticated cellular defense mechanisms to withstand CTL attack. Herein we provide a critical review of the pertinent literature on early and late attack/defense events taking place at the CTL/target cell lytic synapse. We examine the earliest steps of CTL-mediated cytotoxicity (“the poison arrows”) elicited within seconds of CTL/target cell encounter, which face commensurately rapid synaptic repair mechanisms on the tumor cell side, providing the first formidable barrier to CTL attack. We examine how breach of this first defensive barrier unleashes the inextinguishable “Greek fire” in the form of granzymes whose broad cytotoxic potential is linked to activation of cell death executioners, injury of vital organelles, and destruction of intracellular homeostasis. Herein tumor cells deploy slower but no less sophisticated defensive mechanisms in the form of enhanced autophagy, increased reparative capacity, and dysregulation of cell death pathways. We discuss how the newly discovered supra-molecular attack particles (SMAPs, the “scorpion bombs”), seek to overcome the robust defensive mechanisms that confer tumor cell resistance. Finally, we discuss the implications of the aforementioned attack/defense mechanisms on the induction of regulated cell death (RCD), and how different contemporary RCD modalities (including apoptosis, pyroptosis, and ferroptosis) may have profound implications for immunotherapy. Thus, we propose that understanding and targeting multiple steps of the attack/defense process will be instrumental to enhance the efficacy of CTL anti-tumor activity and meet the outstanding challenges in clinical immunotherapy.
Collapse
Affiliation(s)
- Brienne McKenzie
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Roxana Khazen
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Salvatore Valitutti
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France.,Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, Toulouse, France
| |
Collapse
|
19
|
Chen RP, Shinoda K, Rampuria P, Jin F, Bartholomew T, Zhao C, Yang F, Chaparro-Riggers J. Bispecific antibodies for immune cell retargeting against cancer. Expert Opin Biol Ther 2022; 22:965-982. [PMID: 35485219 DOI: 10.1080/14712598.2022.2072209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Following the approval of the T-cell engaging bispecific antibody blinatumomab, immune cell retargeting with bispecific or multispecific antibodies has emerged as a promising cancer immunotherapy strategy, offering alternative mechanisms compared to immune checkpoint blockade. As we gain more understanding of the complex tumor microenvironment, rules and design principles have started to take shape on how to best harness the immune system to achieve optimal anti-tumor activities. AREAS COVERED In the present review, we aim to summarize the most recent advances and challenges in using bispecific antibodies for immune cell retargeting and to provide insights into various aspects of antibody engineering. Discussed herein are studies that highlight the importance of considering antibody engineering parameters, such as binding epitope, affinity, valency, and geometry to maximize the potency and mitigate the toxicity of T cell engagers. Beyond T cell engaging bispecifics, other bispecifics designed to recruit the innate immune system are also covered. EXPERT OPINION Diverse and innovative molecular designs of bispecific/multispecific antibodies have the potential to enhance the efficacy and safety of immune cell retargeting for the treatment of cancer. Whether or not clinical data support these different hypotheses, especially in solid tumor settings, remains to be seen.
Collapse
Affiliation(s)
- Rebecca P Chen
- Pfizer BioMedicine Design, Pfizer Inc, San Diego, CA, USA
| | - Kenta Shinoda
- Pfizer BioMedicine Design, Pfizer Inc, Cambridge, MA, USA
| | | | - Fang Jin
- Pfizer BioMedicine Design, Pfizer Inc, Cambridge, MA, USA
| | | | - Chunxia Zhao
- Pfizer BioMedicine Design, Pfizer Inc, Cambridge, MA, USA
| | - Fan Yang
- Pfizer BioMedicine Design, Pfizer Inc, San Diego, CA, USA
| | | |
Collapse
|
20
|
Perez RK, Gordon MG, Subramaniam M, Kim MC, Hartoularos GC, Targ S, Sun Y, Ogorodnikov A, Bueno R, Lu A, Thompson M, Rappoport N, Dahl A, Lanata CM, Matloubian M, Maliskova L, Kwek SS, Li T, Slyper M, Waldman J, Dionne D, Rozenblatt-Rosen O, Fong L, Dall’Era M, Balliu B, Regev A, Yazdany J, Criswell LA, Zaitlen N, Ye CJ. Single-cell RNA-seq reveals cell type-specific molecular and genetic associations to lupus. Science 2022; 376:eabf1970. [PMID: 35389781 PMCID: PMC9297655 DOI: 10.1126/science.abf1970] [Citation(s) in RCA: 237] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease. Knowledge of circulating immune cell types and states associated with SLE remains incomplete. We profiled more than 1.2 million peripheral blood mononuclear cells (162 cases, 99 controls) with multiplexed single-cell RNA sequencing (mux-seq). Cases exhibited elevated expression of type 1 interferon-stimulated genes (ISGs) in monocytes, reduction of naïve CD4+ T cells that correlated with monocyte ISG expression, and expansion of repertoire-restricted cytotoxic GZMH+ CD8+ T cells. Cell type-specific expression features predicted case-control status and stratified patients into two molecular subtypes. We integrated dense genotyping data to map cell type-specific cis-expression quantitative trait loci and to link SLE-associated variants to cell type-specific expression. These results demonstrate mux-seq as a systematic approach to characterize cellular composition, identify transcriptional signatures, and annotate genetic variants associated with SLE.
Collapse
Affiliation(s)
- Richard K. Perez
- School of Medicine, University of California, San Francisco, CA, USA
| | - M. Grace Gordon
- Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Meena Subramaniam
- Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Min Cheol Kim
- School of Medicine, University of California, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
- UC Berkeley–UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - George C. Hartoularos
- Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Sasha Targ
- School of Medicine, University of California, San Francisco, CA, USA
- Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
| | - Yang Sun
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Anton Ogorodnikov
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Raymund Bueno
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Andrew Lu
- UCLA-Caltech Medical Scientist Training Program, Los Angeles, CA, USA
| | - Mike Thompson
- Department of Computer Science, University of California, Los Angeles, CA, USA
| | - Nadav Rappoport
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Andrew Dahl
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Cristina M. Lanata
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA, USA
| | - Mehrdad Matloubian
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA, USA
| | - Lenka Maliskova
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Serena S. Kwek
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Tony Li
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Michal Slyper
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Julia Waldman
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | | | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Maria Dall’Era
- School of Medicine, University of California, San Francisco, CA, USA
| | - Brunilda Balliu
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jinoos Yazdany
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Lindsey A. Criswell
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA, USA
| | - Noah Zaitlen
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Chun Jimmie Ye
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
21
|
Filali L, Puissegur MP, Cortacero K, Cussat-Blanc S, Khazen R, Van Acker N, Frenois FX, Abreu A, Lamant L, Meyer N, Vergier B, Müller S, McKenzie B, Valitutti S. Ultrarapid lytic granule release from CTLs activates Ca 2+-dependent synaptic resistance pathways in melanoma cells. SCIENCE ADVANCES 2022; 8:eabk3234. [PMID: 35171665 PMCID: PMC8849291 DOI: 10.1126/sciadv.abk3234] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Human cytotoxic T lymphocytes (CTLs) exhibit ultrarapid lytic granule secretion, but whether melanoma cells mobilize defense mechanisms with commensurate rapidity remains unknown. We used single-cell time-lapse microscopy to offer high spatiotemporal resolution analyses of subcellular events in melanoma cells upon CTL attack. Target cell perforation initiated an intracellular Ca2+ wave that propagated outward from the synapse within milliseconds and triggered lysosomal mobilization to the synapse, facilitating membrane repair and conferring resistance to CTL induced cytotoxicity. Inhibition of Ca2+ flux and silencing of synaptotagmin VII limited synaptic lysosomal exposure and enhanced cytotoxicity. Multiplexed immunohistochemistry of patient melanoma nodules combined with automated image analysis showed that melanoma cells facing CD8+ CTLs in the tumor periphery or peritumoral area exhibited significant lysosomal enrichment. Our results identified synaptic Ca2+ entry as the definitive trigger for lysosomal deployment to the synapse upon CTL attack and highlighted an unpredicted defensive topology of lysosome distribution in melanoma nodules.
Collapse
Affiliation(s)
- Liza Filali
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31057 Toulouse, France
| | - Marie-Pierre Puissegur
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31057 Toulouse, France
| | - Kevin Cortacero
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31057 Toulouse, France
| | - Sylvain Cussat-Blanc
- Institut de Recherche en Informatique de Toulouse (IRIT) - University Toulouse Capitole Centre national de la recherche scientifique (CNRS) UMR5505, Artificial and Natural Intelligence Toulouse Institute, Toulouse, France
| | - Roxana Khazen
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31057 Toulouse, France
| | - Nathalie Van Acker
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059 Toulouse, France
| | - François-Xavier Frenois
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059 Toulouse, France
| | - Arnaud Abreu
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059 Toulouse, France
| | - Laurence Lamant
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059 Toulouse, France
| | - Nicolas Meyer
- Department of Dermatology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059 Toulouse, France
| | - Béatrice Vergier
- Service de Pathologie, CHU de Bordeaux, Bordeaux, France
- Equipe INSERM U1053-UMR BaRITOn (Eq 3), Université de Bordeaux, Bordeaux, France
| | - Sabina Müller
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31057 Toulouse, France
| | - Brienne McKenzie
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31057 Toulouse, France
- Corresponding author. (S.V.); (B.M.)
| | - Salvatore Valitutti
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31057 Toulouse, France
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059 Toulouse, France
- Corresponding author. (S.V.); (B.M.)
| |
Collapse
|
22
|
Aschmoneit N, Kühl L, Seifert O, Kontermann RE. Fc-comprising scDb-based trivalent, bispecific T-cell engagers for selective killing of HER3-expressing cancer cells independent of cytokine release. J Immunother Cancer 2021; 9:jitc-2021-003616. [PMID: 34782429 PMCID: PMC8593740 DOI: 10.1136/jitc-2021-003616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2021] [Indexed: 11/10/2022] Open
Abstract
Background Bispecific T-cell engagers are an established therapeutic strategy for the treatment of hematologic malignancies but face several challenges when it comes to their application for the treatment of solid tumors, including on-target off-tumor adverse events. Employing an avidity-mediated specificity gain by introducing an additional binding moiety for the tumor-associated antigen can be achieved using formats with a 2+1 stoichiometry. Methods Besides biochemical characterization and validation of target cell binding to cancer cells with different HER3 expression, we used in vitro co-culture assays with human peripheral blood mononuclear cells (PBMCs) and HER3-expressing target cells to determine T-cell activation, T-cell proliferation and PBMC-mediated cancer cell lysis of HER3-positive cell lines by the trivalent, bispecific antibodies. Results In this study, we developed trivalent, bispecific antibodies comprising a silenced Fc region for T-cell retargeting to HER3-expressing tumor cells, combining a bivalent single-chain diabody (scDb) fused to a first heterodimerizing Fc chain with either an Fab or scFv fused to a second heterodimerizing Fc chain. All these HER3-targeting T-cell engagers comprising two binding sites for HER3 and one binding site for CD3 mediated target cell killing. However, format and orientation of binding sites influenced efficacy of target cell binding, target cell-dependent T-cell activation and T-cell-mediated target cell killing. Beneficial effects were seen when the CD3 binding site was located in the scDb moiety. These molecules showed efficient killing of medium HER3-expressing cancer cells with very low induction of cytokine release, while sparing target cells with low or undetectable HER3 expression. Conclusion Our study demonstrates that these trivalent, bispecific antibodies represent formats with superior interdomain spacing resulting in efficient target cell killing and a potential advantageous safety profile due to very low cytokine release.
Collapse
Affiliation(s)
- Nadine Aschmoneit
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Lennart Kühl
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany .,Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
23
|
Wang X, Sandberg ML, Martin AD, Negri KR, Gabrelow GB, Nampe DP, Wu ML, McElvain ME, Toledo Warshaviak D, Lee WH, Oh J, Daris ME, Chai F, Yao C, Furney J, Pigott C, Kamb A, Xu H. Potent, Selective CARs as Potential T-Cell Therapeutics for HPV-positive Cancers. J Immunother 2021; 44:292-306. [PMID: 34432728 PMCID: PMC8415731 DOI: 10.1097/cji.0000000000000386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 07/08/2021] [Indexed: 11/26/2022]
Abstract
Next-generation T-cell therapies will likely continue to utilize T-cell receptors (TCRs) and chimeric antigen receptors (CARs) because each receptor type has advantages. TCRs often possess exceptional properties even when tested unmodified from patients' T cells. CARs are generally less sensitive, possibly because their ligand-binding domains are grafted from antibodies selected for binding affinity or avidity and not broadly optimized for a functional response. Because of the disconnect between binding and function among these receptor types, the ultimate potential of CARs optimized for sensitivity and selectivity is not clear. Here, we focus on a thoroughly studied immuno-oncology target, the HLA-A*02/HPV-E629-38 complex, and show that CARs can be optimized by a combination of high-throughput binding screens and low-throughput functional assays to have comparable activity to clinical TCRs in acute assays in vitro. These results provide a case study for the challenges and opportunities of optimizing high-performing CARs, especially in the context of targets utilized naturally by TCRs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Julyun Oh
- A2 Biotherapeutics, Agoura Hills, CA
| | | | - Falene Chai
- Innovative Targeting Solutions, Vancouver, BC, Canada
| | - Christine Yao
- Innovative Targeting Solutions, Vancouver, BC, Canada
| | - James Furney
- Innovative Targeting Solutions, Vancouver, BC, Canada
| | - Craig Pigott
- Innovative Targeting Solutions, Vancouver, BC, Canada
| | | | - Han Xu
- A2 Biotherapeutics, Agoura Hills, CA
| |
Collapse
|
24
|
Geuijen C, Tacken P, Wang LC, Klooster R, van Loo PF, Zhou J, Mondal A, Liu YB, Kramer A, Condamine T, Volgina A, Hendriks LJA, van der Maaden H, Rovers E, Engels S, Fransen F, den Blanken-Smit R, Zondag-van der Zande V, Basmeleh A, Bartelink W, Kulkarni A, Marissen W, Huang CY, Hall L, Harvey S, Kim S, Martinez M, O'Brien S, Moon E, Albelda S, Kanellopoulou C, Stewart S, Nastri H, Bakker ABH, Scherle P, Logtenberg T, Hollis G, de Kruif J, Huber R, Mayes PA, Throsby M. A human CD137×PD-L1 bispecific antibody promotes anti-tumor immunity via context-dependent T cell costimulation and checkpoint blockade. Nat Commun 2021; 12:4445. [PMID: 34290245 PMCID: PMC8295259 DOI: 10.1038/s41467-021-24767-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 06/15/2021] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint inhibitors demonstrate clinical activity in many tumor types, however, only a fraction of patients benefit. Combining CD137 agonists with these inhibitors increases anti-tumor activity preclinically, but attempts to translate these observations to the clinic have been hampered by systemic toxicity. Here we describe a human CD137xPD-L1 bispecific antibody, MCLA-145, identified through functional screening of agonist- and immune checkpoint inhibitor arm combinations. MCLA-145 potently activates T cells at sub-nanomolar concentrations, even under suppressive conditions, and enhances T cell priming, differentiation and memory recall responses. In vivo, MCLA-145 anti-tumor activity is superior to immune checkpoint inhibitor comparators and linked to recruitment and intra-tumor expansion of CD8 + T cells. No graft-versus-host-disease is observed in contrast to other antibodies inhibiting the PD-1 and PD-L1 pathway. Non-human primates treated with 100 mg/kg/week of MCLA-145 show no adverse effects. The conditional activation of CD137 signaling by MCLA-145, triggered by neighboring cells expressing >5000 copies of PD-L1, may provide both safety and potency advantages.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Zhou
- Incyte Corporation, Wilmington, DE, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Soyeon Kim
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marina Martinez
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shaun O'Brien
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edmund Moon
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Albelda
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
A scDb-based trivalent bispecific antibody for T-cell-mediated killing of HER3-expressing cancer cells. Sci Rep 2021; 11:13880. [PMID: 34230555 PMCID: PMC8260734 DOI: 10.1038/s41598-021-93351-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/23/2021] [Indexed: 01/12/2023] Open
Abstract
HER3 is a member of the EGF receptor family and elevated expression is associated with cancer progression and therapy resistance. HER3-specific T-cell engagers might be a suitable treatment option to circumvent the limited efficacy observed for HER3-blocking antibodies in clinical trials. In this study, we developed bispecific antibodies for T-cell retargeting to HER3-expressing tumor cells, utilizing either a single-chain diabody format (scDb) with one binding site for HER3 and one for CD3 on T-cells or a trivalent bispecific scDb-scFv fusion protein exhibiting an additional binding site for HER3. The scDb-scFv showed increased binding to HER3-expressing cancer cell lines compared to the scDb and consequently more effective T-cell activation and T-cell proliferation. Furthermore, the bivalent binding mode of the scDb-scFv for HER3 translated into more potent T-cell mediated cancer cell killing, and allowed to discriminate between moderate and low HER3-expressing target cells. Thus, our study demonstrated the applicability of HER3 for T-cell retargeting with bispecific antibodies, even at moderate expression levels, and the increased potency of an avidity-mediated specificity gain, potentially resulting in a wider safety window of bispecific T-cell engaging antibodies targeting HER3.
Collapse
|
26
|
Wang N, Patel H, Schneider IC, Kai X, Varshney AK, Zhou L. An optimal antitumor response by a novel CEA/CD3 bispecific antibody for colorectal cancers. Antib Ther 2021; 4:90-100. [PMID: 34169228 PMCID: PMC8220303 DOI: 10.1093/abt/tbab009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND CD3-based bispecific T cell engagers (bsTCEs) are one of the most promising bispecific antibodies for effective cancer treatments. To elicit target-specific T cell-mediated cytotoxicity, these bsTCEs contain at least one binding unit directed against a tumor antigen and another binding unit targeting CD3 in T cell receptor complex. Development of CD3-based bsTCEs, however, has been severely hampered by dose-limiting toxicities due to cytokine release syndrome. To address this limitation, we developed a novel functionally trivalent T cell engager (t-TCE) antibody containing affinity-reduced CD3 binding unit positioned to ensure monovalent CD3 engagement, in combination with bivalent tumor antigen binding of carcinoembryonic antigen (CEA). METHODS We modeled the variable region of anti-CD3 in the complementarity-determining regions of the heavy chain and obtained CD3 binders with reduced binding affinity. Two optimized versions CEA/CD3-v1 and CEA/CD3-v2 were identified and generated in tetravalent format, characterized and compared in vitro and in vivo for functional activity. RESULTS Our lead candidate, CEA/CD3-v2, demonstrated subnanomolar binding and picomolar potency against a panel of CEA-expressing cancer cell lines. In addition, we detected reduced T cell cytokine release with potent cytotoxic activity. Our t-TCE CEA/CD3-v2 molecule demonstrated strong antitumor effect in a dose-dependent manner in human peripheral blood mononuclear cell (PBMC) xenograft model. Furthermore, combination of CEA/CD3-v2 with atezolizumab provided synergistic antitumor effect. CONCLUSIONS Because of its effective tumor cell killing in vitro and in vivo with reduced cytokine release, CEA/CD3 bsTCE may greatly benefit in CEA-positive cancer immunotherapy.
Collapse
Affiliation(s)
- Ninghai Wang
- Antibody and Cell Therapy Group, Boan Boston LLC, Woburn, MA 01801, USA
| | - Harshal Patel
- Antibody and Cell Therapy Group, Boan Boston LLC, Woburn, MA 01801, USA
| | | | - Xin Kai
- Antibody and Cell Therapy Group, Boan Boston LLC, Woburn, MA 01801, USA
| | - Avanish K Varshney
- Antibody and Cell Therapy Group, Boan Boston LLC, Woburn, MA 01801, USA,To whom correspondence should be addressed. Avanish K. Varshney. ; Li Zhou.
| | - Li Zhou
- Antibody and Cell Therapy Group, Boan Boston LLC, Woburn, MA 01801, USA,To whom correspondence should be addressed. Avanish K. Varshney. ; Li Zhou.
| |
Collapse
|
27
|
Dang K, Castello G, Clarke SC, Li Y, Balasubramani A, Boudreau A, Davison L, Harris KE, Pham D, Sankaran P, Ugamraj HS, Deng R, Kwek S, Starzinski A, Iyer S, van Schooten W, Schellenberger U, Sun W, Trinklein ND, Buelow R, Buelow B, Fong L, Dalvi P. Attenuating CD3 affinity in a PSMAxCD3 bispecific antibody enables killing of prostate tumor cells with reduced cytokine release. J Immunother Cancer 2021; 9:e002488. [PMID: 34088740 PMCID: PMC8183203 DOI: 10.1136/jitc-2021-002488] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Therapeutic options currently available for metastatic castration-resistant prostate cancer (mCRPC) do not extend median overall survival >6 months. Therefore, the development of novel and effective therapies for mCRPC represents an urgent medical need. T cell engagers (TCEs) have emerged as a promising approach for the treatment of mCRPC due to their targeted mechanism of action. However, challenges remain in the clinic due to the limited efficacy of TCEs observed thus far in solid tumors as well as the toxicities associated with cytokine release syndrome (CRS) due to the usage of high-affinity anti-CD3 moieties such as OKT3. METHODS Using genetically engineered transgenic rats (UniRat and OmniFlic) that express fully human IgG antibodies together with an NGS-based antibody discovery pipeline, we developed TNB-585, an anti-CD3xPSMA TCE for the treatment of mCRPC. TNB-585 pairs a tumor-targeting anti-PSMA arm together with a unique, low-affinity anti-CD3 arm in bispecific format. We tested TNB-585 in T cell-redirected cytotoxicity assays against PSMA+ tumor cells in both two-dimensional (2D) cultures and three-dimensional (3D) spheroids as well as against patient-derived prostate tumor cells. Cytokines were measured in culture supernatants to assess the ability of TNB-585 to induce tumor killing with low cytokine release. TNB-585-mediated T cell activation, proliferation, and cytotoxic granule formation were measured to investigate the mechanism of action. Additionally, TNB-585 efficacy was evaluated in vivo against C4-2 tumor-bearing NCG mice. RESULTS In vitro, TNB-585 induced activation and proliferation of human T cells resulting in the killing of PSMA+ prostate tumor cells in both 2D cultures and 3D spheroids with minimal cytokine release and reduced regulatory T cell activation compared with a positive control antibody that contains the same anti-PSMA arm but a higher affinity anti-CD3 arm (comparable with OKT3). In addition, TNB-585 demonstrated potent efficacy against patient-derived prostate tumors ex vivo and induced immune cell infiltration and dose-dependent tumor regression in vivo. CONCLUSIONS Our data suggest that TNB-585, with its low-affinity anti-CD3, may be efficacious while inducing a lower incidence and severity of CRS in patients with prostate cancer compared with TCEs that incorporate high-affinity anti-CD3 domains.
Collapse
Affiliation(s)
| | | | | | - Yuping Li
- Teneobio, Inc, Newark, California, USA
| | | | | | | | | | - Duy Pham
- Teneobio, Inc, Newark, California, USA
| | | | | | - Rong Deng
- Teneobio, Inc, Newark, California, USA
| | - Serena Kwek
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | - Alec Starzinski
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | | | | | | | | | | | | | | | - Lawrence Fong
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
28
|
Crawford A, Chiu D. Targeting Solid Tumors Using CD3 Bispecific Antibodies. Mol Cancer Ther 2021; 20:1350-1358. [PMID: 34045228 DOI: 10.1158/1535-7163.mct-21-0073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/06/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022]
Abstract
Immunotherapies to treat cancer have made tremendous progress over the past decade. In particular, T cell-directed therapies have gained considerable attention with CD3 bispecific antibodies and CAR T cells showing potent responses against hematologic tumors. At present, the ability to adapt these therapeutics to treat solid tumors is less established. Herein, we discuss recent advances in T cell-engaging CD3 bispecific antibodies targeting solid tumors, potential mechanisms of resistance, and future prospects. A better understanding of the mechanisms of immune evasion in solid tumors will enable the development of strategies to overcome this resistance and inform choices of therapeutic combinations.
Collapse
Affiliation(s)
| | - Danica Chiu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| |
Collapse
|
29
|
Zhou Y, Shao N, Bessa de Castro R, Zhang P, Ma Y, Liu X, Huang F, Wang RF, Qin L. Evaluation of Single-Cell Cytokine Secretion and Cell-Cell Interactions with a Hierarchical Loading Microwell Chip. Cell Rep 2021; 31:107574. [PMID: 32348757 PMCID: PMC7583657 DOI: 10.1016/j.celrep.2020.107574] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/21/2020] [Accepted: 04/02/2020] [Indexed: 02/01/2023] Open
Abstract
Comprehensive evaluation of single T cell functions such as cytokine secretion and cytolysis of target cells is greatly needed in adoptive cell therapy (ACT) but has never been fully fulfilled by current approaches. Herein, we develop a hierarchical loading microwell chip (HL-Chip) that aligns multiple cells and functionalized beads in a high-throughput microwell array with single-cell/bead precision based on size differences. We demonstrate the potential of the HL-Chip in evaluating single T cell functions by three applications: high-throughput longitudinal secretory profiling of single T cells, large-scale evaluation of cytolytic activity of single T cells, and integrated T cell-tumor cell interactions. The HL-Chip is a simple and robust technology that constructs arrays of defined cell/object combinations for multiple measurements and material retrieval.
Collapse
Affiliation(s)
- Yufu Zhou
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; The Third Xiangya Hospital, Central South University, Changsha 410008, China; Center for inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Ning Shao
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ricardo Bessa de Castro
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Pengchao Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Yuan Ma
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Xin Liu
- Center for inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine and Norris Comprehensive Cancer Center, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Feizhou Huang
- The Third Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rong-Fu Wang
- Center for inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Pediatrics, Children's Hospital of Los Angeles, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA; Department of Medicine and Norris Comprehensive Cancer Center, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
30
|
Bogen JP, Carrara SC, Fiebig D, Grzeschik J, Hock B, Kolmar H. Design of a Trispecific Checkpoint Inhibitor and Natural Killer Cell Engager Based on a 2 + 1 Common Light Chain Antibody Architecture. Front Immunol 2021; 12:669496. [PMID: 34040611 PMCID: PMC8141644 DOI: 10.3389/fimmu.2021.669496] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
Natural killer cell engagers gained enormous interest in recent years due to their potent anti-tumor activity and favorable safety profile. Simultaneously, chicken-derived antibodies entered clinical studies paving the way for avian-derived therapeutics. In this study, we describe the affinity maturation of a common light chain (cLC)-based, chicken-derived antibody targeting EGFR, followed by utilization of the same light chain for the isolation of CD16a- and PD-L1-specific monoclonal antibodies. The resulting binders target their respective antigen with single-digit nanomolar affinity while blocking the ligand binding of all three respective receptors. Following library-based humanization, bispecific and trispecific variants in a standard 1 + 1 or a 2 + 1 common light chain format were generated, simultaneously targeting EGFR, CD16a, and PD-L1. The trispecific antibody mediated an elevated antibody-dependent cellular cytotoxicity (ADCC) in comparison to the EGFR×CD16a bispecific variant by effectively bridging EGFR/PD-L1 double-positive cancer cells with CD16a-positive effector cells. These findings represent, to our knowledge, the first detailed report on the generation of a trispecific 2 + 1 antibodies exhibiting a common light chain and illustrate synergistic effects of trispecific antigen binding. Overall, this generic procedure paves the way for the engineering of tri- and oligospecific therapeutic antibodies derived from avian immunizations.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody Specificity
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Cell Line, Tumor
- Chickens
- Cytotoxicity, Immunologic/drug effects
- Drug Design
- Epitopes
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/immunology
- ErbB Receptors/metabolism
- Immune Checkpoint Inhibitors/immunology
- Immune Checkpoint Inhibitors/pharmacology
- Immunization
- Immunoglobulin Light Chains/immunology
- Immunoglobulin Light Chains/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Receptors, IgG/antagonists & inhibitors
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Skin Neoplasms/drug therapy
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
Collapse
Affiliation(s)
- Jan P. Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Stefania C. Carrara
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - David Fiebig
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Björn Hock
- Global Pharmaceutical Research and Development, Ferring International Center S.A., Saint-Prex, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
31
|
Chen W, Yang F, Wang C, Narula J, Pascua E, Ni I, Ding S, Deng X, Chu MLH, Pham A, Jiang X, Lindquist KC, Doonan PJ, Van Blarcom T, Yeung YA, Chaparro-Riggers J. One size does not fit all: navigating the multi-dimensional space to optimize T-cell engaging protein therapeutics. MAbs 2021; 13:1871171. [PMID: 33557687 PMCID: PMC7889206 DOI: 10.1080/19420862.2020.1871171] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
T-cell engaging biologics is a class of novel and promising immune-oncology compounds that leverage the immune system to eradicate cancer. Here, we compared and contrasted a bispecific diabody-Fc format, which displays a relatively short antigen-binding arm distance, with our bispecific IgG platform. By generating diverse panels of antigen-expressing cells where B cell maturation antigen is either tethered to the cell membrane or located to the juxtamembrane region and masked by elongated structural spacer units, we presented a systematic approach to investigate the role of antigen epitope location and molecular formats in immunological synapse formation and cytotoxicity. We demonstrated that diabody-Fc is more potent for antigen epitopes located in the membrane distal region, while bispecific IgG is more efficient for membrane-proximal epitopes. Additionally, we explored other parameters, including receptor density, antigen-binding affinity, and kinetics. Our results show that molecular format and antigen epitope location, which jointly determine the intermembrane distance between target cells and T cells, allow decoupling of cytotoxicity and cytokine release, while antigen-binding affinities appear to be positively correlated with both readouts. Our work offers new insight that could potentially lead to a wider therapeutic window for T-cell engaging biologics in general.
Collapse
Affiliation(s)
- Wei Chen
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Asher Bio, Protein Sciences , San Carlos, CA, USA
| | - Fan Yang
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA
| | - Carole Wang
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA
| | - Jatin Narula
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA
| | | | - Irene Ni
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Asher Bio, Protein Sciences , San Carlos, CA, USA
| | - Sheng Ding
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Gilead Sciences, Biology Department , Foster City, CA, USA
| | - Xiaodi Deng
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Dren Bio, Biologics Department , San Carlos, CA, USA
| | - Matthew Ling-Hon Chu
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Tizona Therapeutics, Protein Sciences , Antibody Development & Technical Operations, South San Francisco, CA, USA
| | - Amber Pham
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Arcus Biosciences, Protein Sciences , Hayward, CA, USA
| | - Xiaoyue Jiang
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Nektar Therapeutics, Biologics Analytical Development , San Francisco, CA, USA
| | | | - Patrick J Doonan
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Janssen BioTherapeutics, Janssen Research & Development, LLC , Spring House, PA, USA
| | - Tom Van Blarcom
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Allogene Therapeutics, Protein Engineering , South San Francisco, CA, USA
| | - Yik Andy Yeung
- Pfizer Worldwide R&D , BioMedicine Design, CA, USA.,Asher Bio, Protein Sciences , San Carlos, CA, USA
| | | |
Collapse
|
32
|
Mahnke YD, Devevre E, Baumgaertner P, Matter M, Rufer N, Romero P, Speiser DE. Human melanoma-specific CD8(+) T-cells from metastases are capable of antigen-specific degranulation and cytolysis directly ex vivo. Oncoimmunology 2021; 1:467-530. [PMID: 22754765 PMCID: PMC3382891 DOI: 10.4161/onci.19856] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The relatively low frequencies of tumor Ag-specific T-cells in PBMC and metastases from cancer patients have long precluded the analysis of their direct ex vivo cytolytic capacity. Using a new composite technique that works well with low cell numbers, we aimed at determining the functional competence of melanoma-specific CD8+ T-cells. A multiparameter flow cytometry based technique was applied to assess the cytolytic function, degranulation and IFNγ production by tumor Ag-specific CD8+ T-cells from PBMC and tumor-infiltrated lymph nodes (TILN) of melanoma patients. We found strong cytotoxicity by T-cells not only when they were isolated from PBMC but also from TILN. Cytotoxicity was observed against peptide-pulsed target cells and melanoma cells presenting the naturally processed endogenous antigen. However, unlike their PBMC-derived counterparts, T-cells from TILN produced only minimal amounts of IFNγ, while exhibiting similar levels of degranulation, revealing a critical functional dichotomy in metastatic lesions. Our finding of partial functional impairment fits well with the current knowledge that T-cells from cancer metastases are so-called exhausted, a state of T-cell hyporesponsiveness also found in chronic viral infections. The identification of responsible mechanisms in the tumor microenvironment is important for improving cancer therapies.
Collapse
Affiliation(s)
- Yolanda D Mahnke
- Ludwig Center for Cancer Research; University of Lausanne; Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
33
|
Lafouresse F, Jugele R, Müller S, Doineau M, Duplan-Eche V, Espinosa E, Puisségur MP, Gadat S, Valitutti S. Stochastic asymmetric repartition of lytic machinery in dividing CD8 + T cells generates heterogeneous killing behavior. eLife 2021; 10:62691. [PMID: 33427199 PMCID: PMC7867409 DOI: 10.7554/elife.62691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/08/2021] [Indexed: 12/27/2022] Open
Abstract
Cytotoxic immune cells are endowed with a high degree of heterogeneity in their lytic function, but how this heterogeneity is generated is still an open question. We therefore investigated if human CD8+ T cells could segregate their lytic components during telophase, using imaging flow cytometry, confocal microscopy, and live-cell imaging. We show that CD107a+-intracellular vesicles, perforin, and granzyme B unevenly segregate in a constant fraction of telophasic cells during each division round. Mathematical modeling posits that unequal lytic molecule inheritance by daughter cells results from the random distribution of lytic granules on the two sides of the cleavage furrow. Finally, we establish that the level of lytic compartment in individual cytotoxic T lymphocyte (CTL) dictates CTL killing capacity.
Collapse
Affiliation(s)
- Fanny Lafouresse
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Romain Jugele
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Sabina Müller
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Marine Doineau
- Toulouse School of Economics, CNRS UMR 5314, Université Toulouse 1 Capitole, France and Institut Universitaire de France, Toulouse, France
| | - Valérie Duplan-Eche
- INSERM, UMR1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France
| | - Eric Espinosa
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Marie-Pierre Puisségur
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Sébastien Gadat
- Toulouse School of Economics, CNRS UMR 5314, Université Toulouse 1 Capitole, France and Institut Universitaire de France, Toulouse, France
| | - Salvatore Valitutti
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France.,Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, Toulouse, France
| |
Collapse
|
34
|
Mastrogiovanni M, Juzans M, Alcover A, Di Bartolo V. Coordinating Cytoskeleton and Molecular Traffic in T Cell Migration, Activation, and Effector Functions. Front Cell Dev Biol 2020; 8:591348. [PMID: 33195256 PMCID: PMC7609836 DOI: 10.3389/fcell.2020.591348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/24/2020] [Indexed: 12/28/2022] Open
Abstract
Dynamic localization of receptors and signaling molecules at the plasma membrane and within intracellular vesicular compartments is crucial for T lymphocyte sensing environmental cues, triggering membrane receptors, recruiting signaling molecules, and fine-tuning of intracellular signals. The orchestrated action of actin and microtubule cytoskeleton and intracellular vesicle traffic plays a key role in all these events that together ensure important steps in T cell physiology. These include extravasation and migration through lymphoid and peripheral tissues, T cell interactions with antigen-presenting cells, T cell receptor (TCR) triggering by cognate antigen-major histocompatibility complex (MHC) complexes, immunological synapse formation, cell activation, and effector functions. Cytoskeletal and vesicle traffic dynamics and their interplay are coordinated by a variety of regulatory molecules. Among them, polarity regulators and membrane-cytoskeleton linkers are master controllers of this interplay. Here, we review the various ways the T cell plasma membrane, receptors, and their signaling machinery interplay with the actin and microtubule cytoskeleton and with intracellular vesicular compartments. We highlight the importance of this fine-tuned crosstalk in three key stages of T cell biology involving cell polarization: T cell migration in response to chemokines, immunological synapse formation in response to antigen cues, and effector functions. Finally, we discuss two examples of perturbation of this interplay in pathological settings, such as HIV-1 infection and mutation of the polarity regulator and tumor suppressor adenomatous polyposis coli (Apc) that leads to familial polyposis and colorectal cancer.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Marie Juzans
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Andrés Alcover
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincenzo Di Bartolo
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
35
|
Biolato AM, Filali L, Wurzer H, Hoffmann C, Gargiulo E, Valitutti S, Thomas C. Actin remodeling and vesicular trafficking at the tumor cell side of the immunological synapse direct evasion from cytotoxic lymphocytes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:99-130. [PMID: 33066877 DOI: 10.1016/bs.ircmb.2020.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Andrea Michela Biolato
- Cytoskeleton and Cancer Progression, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Liza Filali
- Cancer Research Center of Toulouse, INSERM, Toulouse, France
| | - Hannah Wurzer
- Cytoskeleton and Cancer Progression, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Céline Hoffmann
- Cytoskeleton and Cancer Progression, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Ernesto Gargiulo
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Salvatore Valitutti
- Cancer Research Center of Toulouse, INSERM, Toulouse, France; Department of Pathology, Institut Universitaire du Cancer-Oncopole, Toulouse, France.
| | - Clément Thomas
- Cytoskeleton and Cancer Progression, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg.
| |
Collapse
|
36
|
Li J, Piskol R, Ybarra R, Chen YJJ, Li J, Slaga D, Hristopoulos M, Clark R, Modrusan Z, Totpal K, Junttila MR, Junttila TT. CD3 bispecific antibody-induced cytokine release is dispensable for cytotoxic T cell activity. Sci Transl Med 2020; 11:11/508/eaax8861. [PMID: 31484792 DOI: 10.1126/scitranslmed.aax8861] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/30/2019] [Indexed: 12/18/2022]
Abstract
T cell-retargeting therapies have transformed the therapeutic landscape of oncology. Regardless of the modality, T cell activating therapies are commonly accompanied by systemic cytokine release, which can progress to deadly cytokine release syndrome (CRS). Because of incomplete mechanistic understanding of the relationship between T cell activation and systemic cytokine release, optimal toxicity management that retains full therapeutic potential remains unclear. Here, we report the cell type-specific cellular mechanisms that link CD3 bispecific antibody-mediated killing to toxic cytokine release. The immunologic cascade is initiated by T cell triggering, whereas monocytes and macrophages are the primary source of systemic toxic cytokine release. We demonstrate that T cell-generated tumor necrosis factor-α (TNF-α) is the primary mechanism mediating monocyte activation and systemic cytokine release after CD3 bispecific treatment. Prevention of TNF-α release is sufficient to impair systemic release of monocyte cytokines without affecting antitumor efficacy. Systemic cytokine release is only observed upon initial exposure to CD3 bispecific antibody not subsequent doses, indicating a biological distinction between doses. Despite impaired cytokine release after second exposure, T cell cytotoxicity remained unaffected, demonstrating that cytolytic activity of T cells can be achieved in the absence of cytokine release. The mechanistic uncoupling of toxic cytokines and T cell cytolytic activity in the context of CD3 bispecifics provides a biological rationale to clinically explore preventative treatment approaches to mitigate toxicity.
Collapse
Affiliation(s)
- Ji Li
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Robert Piskol
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ryan Ybarra
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Jason Li
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Dionysos Slaga
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Robyn Clark
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zora Modrusan
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Klara Totpal
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | |
Collapse
|
37
|
Kist NC, Lambert B, Campbell S, Katzourakis A, Lunn D, Lemey P, Iversen AKN. HIV-1 p24Gag adaptation to modern and archaic HLA-allele frequency differences in ethnic groups contributes to viral subtype diversification. Virus Evol 2020; 6:veaa085. [PMID: 33343925 PMCID: PMC7733611 DOI: 10.1093/ve/veaa085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Pathogen-driven selection and past interbreeding with archaic human lineages have resulted in differences in human leukocyte antigen (HLA)-allele frequencies between modern human populations. Whether or not this variation affects pathogen subtype diversification is unknown. Here we show a strong positive correlation between ethnic diversity in African countries and both human immunodeficiency virus (HIV)-1 p24gag and subtype diversity. We demonstrate that ethnic HLA-allele differences between populations have influenced HIV-1 subtype diversification as the virus adapted to escape common antiviral immune responses. The evolution of HIV Subtype B (HIV-B), which does not appear to be indigenous to Africa, is strongly affected by immune responses associated with Eurasian HLA variants acquired through adaptive introgression from Neanderthals and Denisovans. Furthermore, we show that the increasing and disproportionate number of HIV-infections among African Americans in the USA drive HIV-B evolution towards an Africa-centric HIV-1 state. Similar adaptation of other pathogens to HLA variants common in affected populations is likely.
Collapse
Affiliation(s)
- Nicolaas C Kist
- Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| | - Ben Lambert
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, Medical School Building St Mary’s Campus, Norfolk Place, London W2 1PG, UK
| | - Samuel Campbell
- Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| | - Aris Katzourakis
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| | - Daniel Lunn
- Department of Statistics, University of Oxford, St Giles’, Oxford OX1 3LB, UK
| | - Philippe Lemey
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven - University of Leuven, Leuven B-3000, Belgium
| | - Astrid K N Iversen
- Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
38
|
Lejeune M, Köse MC, Duray E, Einsele H, Beguin Y, Caers J. Bispecific, T-Cell-Recruiting Antibodies in B-Cell Malignancies. Front Immunol 2020; 11:762. [PMID: 32457743 PMCID: PMC7221185 DOI: 10.3389/fimmu.2020.00762] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/03/2020] [Indexed: 12/12/2022] Open
Abstract
Bispecific antibodies (BsAbs) are designed to recognize and bind to two different antigens or epitopes. In the last few decades, BsAbs have been developed within the context of cancer therapies and in particular for the treatment of hematologic B-cell malignancies. To date, more than one hundred different BsAb formats exist, including bispecific T-cell engagers (BiTEs), and new constructs are constantly emerging. Advances in protein engineering have enabled the creation of BsAbs with specific mechanisms of action and clinical applications. Moreover, a better understanding of resistance and evasion mechanisms, as well as advances in the protein engineering and in immunology, will help generating a greater variety of BsAbs to treat various cancer types. This review focuses on T-cell-engaging BsAbs and more precisely on the various BsAb formats currently being studied in the context of B-cell malignancies, on ongoing clinical trials and on the clinical concerns to be taken into account in the development of new BsAbs.
Collapse
Affiliation(s)
- Margaux Lejeune
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Murat Cem Köse
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Elodie Duray
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Hermann Einsele
- Department of Internal Medicine II, University of Würzburg, Würzburg, Germany
| | - Yves Beguin
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium.,Department of Hematology, CHU de Liège, Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium.,Department of Hematology, CHU de Liège, Liège, Belgium
| |
Collapse
|
39
|
Vafa O, Trinklein ND. Perspective: Designing T-Cell Engagers With Better Therapeutic Windows. Front Oncol 2020; 10:446. [PMID: 32351885 PMCID: PMC7174563 DOI: 10.3389/fonc.2020.00446] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/13/2020] [Indexed: 12/22/2022] Open
Abstract
This perspective highlights the history and challenges of developing CD3-based bispecific T-cell engagers (TCEs) as cancer therapeutics as well as considerations and potential strategies for designing the next generation TCE molecules. The goal of this article is to raise awareness of natural T-cell biology and how to best harness the tumor cell killing capacity of cytotoxic T-cells with TCEs. In light of 30 years of concerted efforts to advance TCEs in early clinical development, many of the first-generation bispecific antibodies have exhibited lackluster safety, efficacy, and manufacturability profiles. As of January 2020, blinatumomab remains the only approved TCE. Many of the current set-backs in early clinical trials implicate the high-affinity CD3 binding domains employed and the respective bispecific platforms as potential culprits. The underlying conviction of the authors is that by taking corrective measures, TCEs can transform cancer therapy. Through openness, transparency, and much needed feedback from ongoing clinical studies, the field can continuously improve the design and effectiveness of next generation T-cell redirecting therapeutics.
Collapse
Affiliation(s)
- Omid Vafa
- Teneobio, Inc., Newark, CA, United States
| | | |
Collapse
|
40
|
Hegde PS, Chen DS. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020; 52:17-35. [PMID: 31940268 DOI: 10.1016/j.immuni.2019.12.011] [Citation(s) in RCA: 1292] [Impact Index Per Article: 258.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/01/2019] [Accepted: 12/14/2019] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is a validated and critically important approach for treating patients with cancer. Given the vast research and clinical investigation efforts dedicated to advancing both endogenous and synthetic immunotherapy approaches, there is a need to focus on crucial questions and define roadblocks to the basic understanding and clinical progress. Here, we define ten key challenges facing cancer immunotherapy, which range from lack of confidence in translating pre-clinical findings to identifying optimal combinations of immune-based therapies for any given patient. Addressing these challenges will require the combined efforts of basic researchers and clinicians, and the focusing of resources to accelerate understanding of the complex interactions between cancer and the immune system and the development of improved treatment options for patients with cancer.
Collapse
|
41
|
Saeed MB, Record J, Westerberg LS. Two sides of the coin: Cytoskeletal regulation of immune synapses in cancer and primary immune deficiencies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:1-97. [DOI: 10.1016/bs.ircmb.2020.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Pizzolla A, Wakim LM. Memory T Cell Dynamics in the Lung during Influenza Virus Infection. THE JOURNAL OF IMMUNOLOGY 2019; 202:374-381. [PMID: 30617119 DOI: 10.4049/jimmunol.1800979] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/08/2018] [Indexed: 01/06/2023]
Abstract
Influenza A virus is highly contagious, infecting 5-15% of the global population every year. It causes significant morbidity and mortality, particularly among immunocompromised and at-risk individuals. Influenza virus is constantly evolving, undergoing continuous, rapid, and unpredictable mutation, giving rise to novel viruses that can escape the humoral immunity generated by current influenza virus vaccines. Growing evidence indicates that influenza-specific T cells resident along the respiratory tract are highly effective at providing potent and rapid protection against this inhaled pathogen. As these T cells recognize fragments of the virus that are highly conserved and less prone to mutation, they have the potential to provide cross-strain protection against a wide breadth of influenza viruses, including newly emerging strains. In this review, we will discuss how influenza-specific memory T cells in the lung are established and maintained and how we can harness this knowledge to design broadly protective influenza A virus vaccines.
Collapse
Affiliation(s)
- Angela Pizzolla
- Department of Microbiology and Immunology, University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| |
Collapse
|
43
|
Khazen R, Müller S, Lafouresse F, Valitutti S, Cussat-Blanc S. Sequential adjustment of cytotoxic T lymphocyte densities improves efficacy in controlling tumor growth. Sci Rep 2019; 9:12308. [PMID: 31444380 PMCID: PMC6707257 DOI: 10.1038/s41598-019-48711-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/09/2019] [Indexed: 12/30/2022] Open
Abstract
Understanding the human cytotoxic T lymphocyte (CTL) biology is crucial to develop novel strategies aiming at maximizing their lytic capacity against cancer cells. Here we introduce an agent-based model, calibrated on population-scale experimental data that allows quantifying human CTL per capita killing. Our model highlights higher individual CTL killing capacity at lower CTL densities and fits experimental data of human melanoma cell killing. The model allows extending the analysis over prolonged time frames, difficult to investigate experimentally, and reveals that initial high CTL densities hamper efficacy to control melanoma growth. Computational analysis forecasts that sequential addition of fresh CTL cohorts improves tumor growth control. In vivo experimental data, obtained in a mouse melanoma model, confirm this prediction. Taken together, our results unveil the impact that sequential adjustment of cellular densities has on enhancing CTL efficacy over long-term confrontation with tumor cells. In perspective, they can be instrumental to refine CTL-based therapeutic strategies aiming at controlling tumor growth.
Collapse
Affiliation(s)
- Roxana Khazen
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France.,INSERM U1223, Dynamics of Immune Responses Unit, Institut Pasteur, 75015, Paris, France
| | - Sabina Müller
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France
| | - Fanny Lafouresse
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France
| | - Salvatore Valitutti
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France. .,Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059, Toulouse, France.
| | - Sylvain Cussat-Blanc
- Institute of Advanced Technologies in Living Sciences, CNRS - USR3505, Toulouse, France.,University of Toulouse, Institute of Research in Informatics of Toulouse, CNRS - UMR5505, Toulouse, France
| |
Collapse
|
44
|
Super-resolution microscopy reveals ultra-low CD19 expression on myeloma cells that triggers elimination by CD19 CAR-T. Nat Commun 2019; 10:3137. [PMID: 31316055 PMCID: PMC6637169 DOI: 10.1038/s41467-019-10948-w] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 06/10/2019] [Indexed: 01/27/2023] Open
Abstract
Immunotherapy with chimeric antigen receptor-engineered T-cells (CAR-T) is under investigation in multiple myeloma. There are reports of myeloma remission after CD19 CAR-T therapy, although CD19 is hardly detectable on myeloma cells by flow cytometry (FC). We apply single molecule-sensitive direct stochastic optical reconstruction microscopy (dSTORM), and demonstrate CD19 expression on a fraction of myeloma cells (10.3–80%) in 10 out of 14 patients (density: 13–5,000 molecules per cell). In contrast, FC detects CD19 in only 2 of these 10 patients, on a smaller fraction of cells. Treatment with CD19 CAR-T in vitro results in elimination of CD19-positive myeloma cells, including those with <100 CD19 molecules per cell. Similar data are obtained by dSTORM analyses of CD20 expression on myeloma cells and CD20 CAR-T. These data establish a sensitivity threshold for CAR-T and illustrate how super-resolution microscopy can guide patient selection in immunotherapy to exploit ultra-low density antigens. CD19 CAR-T cells have achieved some success in treating myeloma patients despite the limited detection of the CD19 antigen. Here, the authors show using dSTORM that 10/14 myeloma samples studied express ultra-low levels of CD19, which are sufficient for engaging CAR-T cells in vitro.
Collapse
|
45
|
Trinklein ND, Pham D, Schellenberger U, Buelow B, Boudreau A, Choudhry P, Clarke SC, Dang K, Harris KE, Iyer S, Jorgensen B, Pratap PP, Rangaswamy US, Ugamraj HS, Vafa O, Wiita AP, van Schooten W, Buelow R, Force Aldred S. Efficient tumor killing and minimal cytokine release with novel T-cell agonist bispecific antibodies. MAbs 2019; 11:639-652. [PMID: 30698484 PMCID: PMC6601548 DOI: 10.1080/19420862.2019.1574521] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
T-cell-recruiting bispecific antibodies (T-BsAbs) have shown potent tumor killing activity in humans, but cytokine release-related toxicities have affected their clinical utility. The use of novel anti-CD3 binding domains with more favorable properties could aid in the creation of T-BsAbs with improved therapeutic windows. Using a sequence-based discovery platform, we identified new anti-CD3 antibodies from humanized rats that bind to multiple epitopes and elicit varying levels of T-cell activation. In T-BsAb format, 12 different anti-CD3 arms induce equivalent levels of tumor cell lysis by primary T-cells, but potency varies by a thousand-fold. Our lead CD3-targeting arm stimulates very low levels of cytokine release, but drives robust tumor antigen-specific killing in vitro and in a mouse xenograft model. This new CD3-targeting antibody underpins a next-generation T-BsAb platform in which potent cytotoxicity is uncoupled from high levels of cytokine release, which may lead to a wider therapeutic window in the clinic.
Collapse
Affiliation(s)
| | - Duy Pham
- a Teneobio, Inc ., Menlo Park , CA , USA
| | | | - Ben Buelow
- a Teneobio, Inc ., Menlo Park , CA , USA
| | | | - Priya Choudhry
- b Department of Laboratory Medicine , University of California , San Francisco , CA , USA
| | | | - Kevin Dang
- a Teneobio, Inc ., Menlo Park , CA , USA
| | | | | | | | | | | | | | - Omid Vafa
- a Teneobio, Inc ., Menlo Park , CA , USA
| | - Arun P Wiita
- b Department of Laboratory Medicine , University of California , San Francisco , CA , USA
| | | | | | | |
Collapse
|
46
|
Tilburgs T, Meissner TB, Ferreira LMR, Mulder A, Musunuru K, Ye J, Strominger JL. NLRP2 is a suppressor of NF-ƙB signaling and HLA-C expression in human trophoblasts†,‡. Biol Reprod 2018; 96:831-842. [PMID: 28340094 DOI: 10.1093/biolre/iox009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/28/2017] [Indexed: 01/06/2023] Open
Abstract
During pregnancy, fetal extravillous trophoblasts (EVT) play a key role in the regulation of maternal T cell and NK cell responses. EVT display a unique combination of human leukocyte antigens (HLA); EVT do not express HLA-A and HLA-B, but do express HLA-C, HLA-E, and HLA-G. The mechanisms establishing this unique HLA expression pattern have not been fully elucidated. The major histocompatibility complex (MHC) class I and class II transcriptional activators NLRC5 and CIITA are expressed neither by EVT nor by the EVT model cell line JEG3, which has an MHC expression pattern identical to that of EVT. Therefore, other MHC regulators must be present to control HLA-C, HLA-E, and HLA-G expression in these cells. CIITA and NLRC5 are both members of the nucleotide-binding domain, leucine-rich repeat (NLR) family of proteins. Another member of this family, NLRP2, is highly expressed by EVT and JEG3, but not in maternal decidual stromal cells. In this study, transcription activator-like effector nuclease technology was used to delete NLRP2 in JEG3. Furthermore, lentiviral delivery of shRNA was used to knockdown NLRP2 in JEG3 and primary EVT. Upon NLRP2 deletion, Tumor Necrosis Factor-α (TNFα)-induced phosphorylation of NF-KB p65 increased in JEG3 and EVT, and more surprisingly a significant increase in constitutive HLA-C expression was observed in JEG3. These data suggest a broader role for NLR family members in the regulation of MHC expression during inflammation, thus forming a bridge between innate and adaptive immune responses. As suppressor of proinflammatory responses, NLRP2 may contribute to preventing unwanted antifetal responses.
Collapse
Affiliation(s)
- Tamara Tilburgs
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Torsten B Meissner
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Leonardo M R Ferreira
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Arend Mulder
- Department of Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Kiran Musunuru
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Junqiang Ye
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA.,Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York, USA
| | - Jack L Strominger
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
47
|
Kabanova A, Zurli V, Baldari CT. Signals Controlling Lytic Granule Polarization at the Cytotoxic Immune Synapse. Front Immunol 2018. [PMID: 29515593 PMCID: PMC5826174 DOI: 10.3389/fimmu.2018.00307] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cytotoxic immunity relies on specialized effector T cells, the cytotoxic T cells, which are endowed with specialized cytolytic machinery that permits them to induce death of their targets. Upon recognition of a target cell, cytotoxic T cells form a lytic immune synapse and by docking the microtubule-organizing center at the synaptic membrane get prepared to deliver a lethal hit of enzymes contained in lytic granules. New insights suggest that the directionality of lytic granule trafficking along the microtubules represents a fine means to tune the functional outcome of the encounter between a T cell and its target. Thus, mechanisms regulating the directionality of granule transport may have a major impact in settings characterized by evasion from the cytotoxic response, such as chronic infection and cancer. Here, we review our current knowledge on the signaling pathways implicated in the polarized trafficking at the immune synapse of cytotoxic T cells, complementing it with information on the regulation of this process in natural killer cells. Furthermore, we highlight some of the parameters which we consider critical in studying the polarized trafficking of lytic granules, including the use of freshly isolated cytotoxic T cells, and discuss some of the major open questions.
Collapse
Affiliation(s)
- Anna Kabanova
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Vanessa Zurli
- Department of Life Sciences, University of Siena, Siena, Italy
| | | |
Collapse
|
48
|
Abstract
Harnessing the power of the human immune system to treat cancer is the essence of immunotherapy. Monoclonal antibodies engage the innate immune system to destroy targeted cells. For the last 30years, antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity have been the main mechanisms of anti-tumor action of unconjugated antibody drugs. Efforts to exploit the potentials of other immune cells, in particular T cells, culminated in the recent approval of two T cell engaging bispecific antibody (T-BsAb) drugs, thereby stimulating new efforts to accelerate similar platforms through preclinical and clinical trials. In this review, we have compiled the worldwide effort in exploring T cell engaging bispecific antibodies. Our special emphasis is on the lessons learned, with the hope to derive insights in this fast evolving field with tremendous clinical potential.
Collapse
Affiliation(s)
- Z Wu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - N V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
49
|
Yong CSM, John LB, Devaud C, Prince MH, Johnstone RW, Trapani JA, Darcy PK, Kershaw MH. A role for multiple chimeric antigen receptor-expressing leukocytes in antigen-specific responses to cancer. Oncotarget 2018; 7:34582-98. [PMID: 27153556 PMCID: PMC5085178 DOI: 10.18632/oncotarget.9149] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/16/2016] [Indexed: 12/18/2022] Open
Abstract
While adoptive immunotherapy using chimeric antigen receptor (CAR)-modified T cells can induce remission of some tumors, the role of other CAR-modified leukocytes is not well characterized. In this study, we characterize the function of leukocytes including natural killer (NK) cells, macrophages and CAR T cells from transgenic mice expressing a CAR under the control of the pan-hematopoietic promoter, vav, and determine the ability of these mice to respond to ERB expressing tumors. We demonstrate the anti-tumor functions of leukocytes, including antigen specific cytotoxicity and cytokine secretion. The adoptive transfer of CAR T cells provided a greater survival advantage in the E0771ERB tumor model than their wildtype (WT) counterparts. In addition, CAR NK cells and CAR T cells also mediated increased survival in the RMAERB tumor model. When challenged with Her2 expressing tumors, F38 mice were shown to mount an effective immune response, resulting in tumor rejection and long-term survival. This was shown to be predominantly dependent on both CD8+ T cells and NK cells. However, macrophages and CD4+ T cells were also shown to contribute to this response. Overall, this study highlights the use of the vav-CAR mouse model as a unique tool to determine the anti-tumor function of various immune subsets, either alone or when acting alongside CAR T cells in adoptive immunotherapy.
Collapse
Affiliation(s)
- Carmen S M Yong
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Liza B John
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Christel Devaud
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Institut de Recherche en Santé Digestive, Université de Toulouse, INPT, INRA, INSERM UMR1220, UPS, France
| | - Miles H Prince
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Ricky W Johnstone
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Joseph A Trapani
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Phillip K Darcy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Immunology, Monash University, Prahran Victoria, Australia
| | - Michael H Kershaw
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Immunology, Monash University, Prahran Victoria, Australia
| |
Collapse
|
50
|
Walker AJ, Majzner RG, Zhang L, Wanhainen K, Long AH, Nguyen SM, Lopomo P, Vigny M, Fry TJ, Orentas RJ, Mackall CL. Tumor Antigen and Receptor Densities Regulate Efficacy of a Chimeric Antigen Receptor Targeting Anaplastic Lymphoma Kinase. Mol Ther 2017; 25:2189-2201. [PMID: 28676342 DOI: 10.1016/j.ymthe.2017.06.008] [Citation(s) in RCA: 271] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 06/06/2017] [Accepted: 06/09/2017] [Indexed: 12/11/2022] Open
Abstract
We explored the utility of targeting anaplastic lymphoma kinase (ALK), a cell surface receptor overexpressed on pediatric solid tumors, using chimeric antigen receptor (CAR)-based immunotherapy. T cells expressing a CAR incorporating the single-chain variable fragment sequence of the ALK48 mAb linked to a 4-1BB-CD3ζ signaling domain lysed ALK-expressing tumor lines and produced interferon-gamma upon antigen stimulation but had limited anti-tumor efficacy in two xenograft models of human neuroblastoma. Further exploration demonstrated that cytokine production was highly dependent upon ALK target density and that target density of ALK on neuroblastoma cell lines was insufficient for maximal activation of CAR T cells. In addition, ALK CAR T cells demonstrated rapid and complete antigen-induced loss of receptor from the T cell surface via internalization. Using a model that simultaneously modulated antigen density and CAR expression, we demonstrated that CAR functionality is regulated by target antigen and CAR density and that low expression of either contributes to limited anti-tumor efficacy of the ALK CAR. These data suggest that stoichiometric relationships between CAR receptors and target antigens may significantly impact the anti-tumor efficacy of CAR T cells and that manipulation of these parameters could allow precise tuning of CAR T cell activity.
Collapse
Affiliation(s)
- Alec J Walker
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Robbie G Majzner
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ling Zhang
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Kelsey Wanhainen
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Adrienne H Long
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Sang M Nguyen
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Paola Lopomo
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Marc Vigny
- INSERM/UPMC, Institut du Fer à Moulin, 75005 Paris, France
| | - Terry J Fry
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Rimas J Orentas
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Crystal L Mackall
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|