EHA Library - The official digital education library of European Hematology Association (EHA)

LAG-3, TIM-3 OR 2B4 DISRUPTION DIFFERENTIALLY REGULATE THE ANTI-TUMOR RESPONSE OF TCR GENE EDITED HUMAN MEMORY STEM T CELLS
Author(s): ,
Beatrice Claudia Cianciotti
Affiliations:
Division of Immunology, Transplantation and Infectous Diseases - Experimental Hematology Unit,San Raffaele Scientific Institute,Milano,Italy
,
Zulma Irene Magnani
Affiliations:
Division of Immunology, Transplantation and Infectous Diseases - Experimental Hematology Unit,San Raffaele Scientific Institute,Milano,Italy
,
Barbara Camisa
Affiliations:
Division of Immunology, Transplantation and Infectous Diseases - Experimental Hematology Unit,San Raffaele Scientific Institute,Milano,Italy
,
Alessia Potenza
Affiliations:
Division of Immunology, Transplantation and Infectous Diseases - Experimental Hematology Unit,San Raffaele Scientific Institute,Milano,Italy
,
Valentina Vavassori
Affiliations:
Gene transfer technology and new gene therapy strategies Unit, TIGET,San Raffaele Scientific Institute,Milano,Italy
,
Luigi Naldini
Affiliations:
TIGET,San Raffaele Scientific Institute,Milano,Italy
,
Fabio Ciceri
Affiliations:
Hematology and Bone Marrow Transplantation Unit,San Raffaele Scientific Institute, Università Vita-Salute San Raffaele,Milano,Italy
,
Pietro Genovese
Affiliations:
Gene transfer technology and new gene therapy strategies Unit, TIGET,San Raffaele Scientific Institute,Milano,Italy
,
Eliana Ruggiero
Affiliations:
Division of Immunology, Transplantation and Infectous Diseases - Experimental Hematology Unit,San Raffaele Scientific Institute,Milano,Italy
Chiara Bonini
Affiliations:
Division of Immunology, Transplantation and Infectous Diseases - Experimental Hematology Unit,San Raffaele Scientific Institute,Milano,Italy
EHA Library. Claudia Cianciotti B. 06/09/21; 324660; S252
Beatrice Claudia Cianciotti
Beatrice Claudia Cianciotti
Contributions
Abstract
Presentation during EHA2021: All Oral presentations will be made available as of Friday, June 11, 2021 (09:00 CEST) and will be accessible for on-demand viewing until August 15, 2021 on the Virtual Congress platform.

Abstract: S252

Type: Oral Presentation

Session title: Cellular immunotherapy and gene therapy - Experimental

Background
Adoptive immunotherapy with T lymphocytes genetically engineered to overexpress a tumor-specific T cell receptor represents a novel immunotherapeutic option for cancer patients. However, the lack of long-term persistence of infused tumor-reactive T cells and the immunosuppressive tumor microenvironment represent major drawbacks of ACT. Specifically, chronically stimulated tumor-specific lymphocytes become exhausted, a dysfunctional status characterized by loss of effector functions. Exhausted T cells overexpress several inhibitory receptors (IRs), such as PD-1, CTLA-4, Lag-3, Tim-3 and 2B4. The use of monoclonal antibodies targeting IRs (e.g. anti-CTLA-4 and anti-PD-1), can revert T cell exhaustion but proved effective only in a fraction of patients. Also, autoimmune-related adverse events often occur after immune checkpoint blockade, mainly due to the unleashing of autoreactive clones. 

Aims
we aim to generate innovative cell therapy products, endowed with long-term persisting capacity and resistance to inhibitory signals and to investigate how the disruption of selected IRs could modulate the resistance to exhaustion in multiple myeloma models. 

Methods
CRISPR/Cas9 reagents were used to simultaneously disrupt TIM-3, LAG-3 or 2B4 gene and the TCR alpha chain constant region (TRAC) gene. The frequency of NHEJ was assessed with ddPCR. TRAC-KO-IR-KO T cells were transduced with a lentiviral vector encoding for an HLA-A2-restricted NY-ESO-1-specific TCR. T cells were activated and kept in culture with a protocol to expand long-living early differentiated stem cell memory (TSCM) and central memory (TCM) cells (Cieri et al., Blood 2013). TCR-edited-IR-KO cells and TCR-edited-IR-competent (TCR-edited-IRCOMP) cells were challenged in vitro and in vivo with HLA-A2posNY-ESO-1pos multiple myeloma cell lines. HLA-A2neg NYESO-1neg cell lines were used as control.  

Results
We efficiently inactivated the endogenous TCR (>98% of NHEJ) and more than 80% of gene disruption was also obtained at LAG-3, Tim-3 or 2B4 locus. TCR disrupted-IR-KO cells were efficiently transduced (> 50%) with the NY-ESO1157-165-specific TCR, thus obtaining TCR-edited-IR-KO T cells or TCR-edited-IRCOMPT cells. Importantly, TCR-edited-IR-KO T cells retain their expansion capacity and an early differentiated TSCM/TCM phenotype. 


Upon chronic stimulation with multiple myeloma cells, TCR-edited-Tim-3-KO and TCR-edited-2B4-KO T cells retained a higher degranulation capacity compared to TCR-edited-LAG-3 KO and TCR-edited-IRCOMP T cells, while TCR-edited-LAG-3-KO T cells showed a limited upregulation of additional inhibitory receptors. 


In a preclinical model of aggressive multiple myeloma, TCR-edited-Tim3-KO and TCR-edited-2B4-KO T cells caused higher immunological pressure and were enriched in a higher proportion of highly activated T cells compared to TCR-edited-IRCOMP-treated mice. In a similar pre-clinical multiple myeloma model, TCR-edited-LAG-3-KO, but not TCR-edited-IRCOMP, T cells showed a reduced expression of additional IRs and prevented the engraftment of the second infusion of tumor cells, thus indicating an enhanced T cell fitness and resistance to exhaustion of TCR-edited-LAG-3-KO T cells.

Conclusion
our results show that by combining the versatility of multiplex gene editing by CRISPR/Cas9 with culture conditions designed to engineer TSCM cells, we can generate innovative tumor-specific cellular products redirected against tumor antigens and resistant to different inhibitory signals.

Keyword(s): Gene therapy, Immunotherapy

Presentation during EHA2021: All Oral presentations will be made available as of Friday, June 11, 2021 (09:00 CEST) and will be accessible for on-demand viewing until August 15, 2021 on the Virtual Congress platform.

Abstract: S252

Type: Oral Presentation

Session title: Cellular immunotherapy and gene therapy - Experimental

Background
Adoptive immunotherapy with T lymphocytes genetically engineered to overexpress a tumor-specific T cell receptor represents a novel immunotherapeutic option for cancer patients. However, the lack of long-term persistence of infused tumor-reactive T cells and the immunosuppressive tumor microenvironment represent major drawbacks of ACT. Specifically, chronically stimulated tumor-specific lymphocytes become exhausted, a dysfunctional status characterized by loss of effector functions. Exhausted T cells overexpress several inhibitory receptors (IRs), such as PD-1, CTLA-4, Lag-3, Tim-3 and 2B4. The use of monoclonal antibodies targeting IRs (e.g. anti-CTLA-4 and anti-PD-1), can revert T cell exhaustion but proved effective only in a fraction of patients. Also, autoimmune-related adverse events often occur after immune checkpoint blockade, mainly due to the unleashing of autoreactive clones. 

Aims
we aim to generate innovative cell therapy products, endowed with long-term persisting capacity and resistance to inhibitory signals and to investigate how the disruption of selected IRs could modulate the resistance to exhaustion in multiple myeloma models. 

Methods
CRISPR/Cas9 reagents were used to simultaneously disrupt TIM-3, LAG-3 or 2B4 gene and the TCR alpha chain constant region (TRAC) gene. The frequency of NHEJ was assessed with ddPCR. TRAC-KO-IR-KO T cells were transduced with a lentiviral vector encoding for an HLA-A2-restricted NY-ESO-1-specific TCR. T cells were activated and kept in culture with a protocol to expand long-living early differentiated stem cell memory (TSCM) and central memory (TCM) cells (Cieri et al., Blood 2013). TCR-edited-IR-KO cells and TCR-edited-IR-competent (TCR-edited-IRCOMP) cells were challenged in vitro and in vivo with HLA-A2posNY-ESO-1pos multiple myeloma cell lines. HLA-A2neg NYESO-1neg cell lines were used as control.  

Results
We efficiently inactivated the endogenous TCR (>98% of NHEJ) and more than 80% of gene disruption was also obtained at LAG-3, Tim-3 or 2B4 locus. TCR disrupted-IR-KO cells were efficiently transduced (> 50%) with the NY-ESO1157-165-specific TCR, thus obtaining TCR-edited-IR-KO T cells or TCR-edited-IRCOMPT cells. Importantly, TCR-edited-IR-KO T cells retain their expansion capacity and an early differentiated TSCM/TCM phenotype. 


Upon chronic stimulation with multiple myeloma cells, TCR-edited-Tim-3-KO and TCR-edited-2B4-KO T cells retained a higher degranulation capacity compared to TCR-edited-LAG-3 KO and TCR-edited-IRCOMP T cells, while TCR-edited-LAG-3-KO T cells showed a limited upregulation of additional inhibitory receptors. 


In a preclinical model of aggressive multiple myeloma, TCR-edited-Tim3-KO and TCR-edited-2B4-KO T cells caused higher immunological pressure and were enriched in a higher proportion of highly activated T cells compared to TCR-edited-IRCOMP-treated mice. In a similar pre-clinical multiple myeloma model, TCR-edited-LAG-3-KO, but not TCR-edited-IRCOMP, T cells showed a reduced expression of additional IRs and prevented the engraftment of the second infusion of tumor cells, thus indicating an enhanced T cell fitness and resistance to exhaustion of TCR-edited-LAG-3-KO T cells.

Conclusion
our results show that by combining the versatility of multiplex gene editing by CRISPR/Cas9 with culture conditions designed to engineer TSCM cells, we can generate innovative tumor-specific cellular products redirected against tumor antigens and resistant to different inhibitory signals.

Keyword(s): Gene therapy, Immunotherapy

By clicking “Accept Terms & all Cookies” or by continuing to browse, you agree to the storing of third-party cookies on your device to enhance your user experience and agree to the user terms and conditions of this learning management system (LMS).

Cookie Settings
Accept Terms & all Cookies