-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

665 Bortezomib Induces Anti-Multiple Myeloma Immune Response Mediated By Cgas/Sting Pathway Activation, Type I Interferon Secretion, and Immunogenic Cell Death: Clinical Application

Program: Oral and Poster Abstracts
Type: Oral
Session: 652. Myeloma: Pathophysiology and Pre-Clinical Studies, excluding Therapy
Hematology Disease Topics & Pathways:
multiple myeloma, Diseases, Therapies, Combinations, Biological Processes, Plasma Cell Disorders, Lymphoid Malignancies, Clinically relevant, immune mechanism, microenvironment
Monday, December 7, 2020: 11:30 AM

Annamaria Gulla, MD1, Eugenio Morelli, MD2,3, Mehmet Kemal Samur, PhD4*, Cirino Botta, MD, PhD5, Teru Hideshima, MD, PhD6, Giada Bianchi, MD3,7,8, Mariateresa Fulciniti, PhD9, Stefano Malvestiti, MD10*, Rao Prabhala9,11, Srikanth Talluri, PhD12,13*, Yu-Tzu Tai, PhD14, Kenneth Wen15*, Paul G. Richardson, MD12,16, Ruben D. Carrasco, MD, PhD3,17,18*, Dharminder Chauhan, PhD19, Nikhil C. Munshi, MD3,4,11 and Kenneth Anderson, MD20,21

1Dana Farber Cancer Institute, Boston, MA
2Department of Medical Oncology, DANA FARBER CANCER INSTITUTE, Boston, MA
3Harvard Medical School, Boston, MA
4Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
5Hematology Unit, "Annunziata" Hospital of Cosenza, Cosenza, Italy
6Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
7Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
8Division of hematology, Brigham and Women's Hospital, Boston
9The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
10Department of Pediatric Hematology and Oncology, Freiburg University Hospital, Freiburg, Germany
11Boston VA Healthcare System, West Roxbury, MA
12Dana-Farber Cancer Institute, Boston, MA
13VA Boston Healthcare System, Boston, MA
14Department of Medical Oncology, Dana-Farber Cancer Inst., Boston, MA
15Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Boston, MA
16Medical Oncology, Harvard Medical School, Boston, MA
17Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
18Dept. of Pathology, Brigham and Women's Hospital, Boston, MA
19Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
20LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
21Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA

Immune escape underlies progression of disease and resistance to therapy in MM, and dysfunction of both innate and adaptive immunity highlight the urgent need for scientifically-informed strategies to restore anti-MM immunity and improve patient outcome. Here we demonstrate that proteasome inhibitor bortezomib (BTZ) modulates the immunosuppressive bone marrow milieu by inducing immunogenic cell death (ICD) and a type-I interferon (IFN) response which contribute to its clinical efficacy. Moreover, we identified STING as a mediator of its anti-MM immune response, and provide the preclinical rationale for clinical trials of BTZ-STING agonist combination therapy.

We first show that BTZ induces hallmarks of ICD in both human and murine MM cell lines, including exposure of endoplasmic reticulum protein calreticulin (CALR), that functions as an “eat me signal”. Specifically, co-culture with BTZ-treated MM cells induced functional maturation of dendritic cells (DCs) and enhanced uptake of BTZ-treated MM cells, assessed by flow cytometry and confocal microscopy. Notably, these functional sequelae were abrogated when DCs were co-cultured with CALRKO MM cells; and add-back experiments by stable overexpression of CALR in KO clones confirmed the specific role of CALR in BTZ-induced immunogenicity. We next validated these findings in 2 in vivo syngeneic models. First, we observed that anti-MM activity of BTZ resulted in more potent murine 5TGM1 tumor cell shrinkage in immunocompetent than immunodeficient hosts, an effect mediated by ICD induction since it was abrogated in immunocompetent mice bearing CALRKO tumors. Second, in vitro BTZ-treated 5TGM1 cells were used as a vaccine to induce a protective anti-MM immune response: vaccination protected against tumor growth upon rechallenge with 5TGM1 cells; conversely, palpable tumors were detected in non-vaccinated mice by 1 week. Moreover, vaccination with BTZ-treated CALRKO 5TGM1 cells abrogated the protective effect of BTZ since only 50% mice were tumor free by 30 days. Consistently, ELISPOT assay on mice splenocytes confirmed that vaccination with BTZ-treated 5TGM1 cells triggered a MM-specific T cell response.

Next, we performed RNAseq analysis of BTZ-treated vs untreated tumors from both CALRWT and CALRKO cells growing in immunocompetent mice; and then carried out an integrative analysis of RNAseq data from clinically-annotated MM patients (n=327) uniformly treated with BTZ-based regimens (IFM/DFCI 2009). We identified a specific ICD signature induced by BTZ only in CALRWT tumors in mice; and importantly, we found that increased expression of the human orthologs of this ICD signature was strongly and positively correlated with clinical outcome (overall survival (OS), p value=0.01). Moreover, the predictive value of this signature was confirmed in an independent dataset of BTZ-treated patients (GSE9782) (OS p value=0.024). Gene ontology analysis of the ICD signature genes revealed enrichment in inflammatory response pathways, including IFN stimulated genes (ISGs). Using RNAseq analysis and qRT-PCR validation, we showed that MM cells demonstrate a type-I IFN response after BTZ treatment which contributes to its in vivo efficacy, since neutralization of type-I IFNs signaling in both MM and host cells with a type-I IFNs receptor 1 (IFNAR) blocking antibody significantly reduced BTZ anti-MM activity. Finally, we showed that BTZ increases genomic instability/micronuclei formation in MM cells and activates innate cGAS/STING immune pathway, thereby stimulating a type-I IFN response. Conversely, STINGKO in MM cells abrogated this effect and decreased BTZ-triggered anti-MM T cell response. Moreover, expression of the ISGs included in ICD signature was positively correlated with STING expression in MM patients. Importantly, pharmacological activation of STING with STING agonist induced potent anti-MM activity in vivo; and combination of STING agonist with BTZ further potentiated this in vivo anti-MM response, with increased T cell infiltration into retrieved tumors, evidenced by IHC analysis.

In conclusion, our studies delineate a novel mechanism whereby BTZ triggers anti-MM immune responses, and show that STING agonists can enhance this response. These findings provide the framework for clinical evaluation of STING agonists in combination with BTZ to induce potent anti-MM immune responses and thereby improve patient outcome.

Disclosures: Fulciniti: NIH: Research Funding. Richardson: Celgene/BMS, Oncopeptides, Takeda, Karyopharm: Research Funding. Chauhan: Oncopeptide AB: Consultancy; consultant to Stemline Therapeutics, Inc., and Equity owner in C4 Therapeutics.: Consultancy, Other: Equity owner in C4 Therapeutics.. Munshi: Karyopharm: Consultancy; Takeda: Consultancy; AbbVie: Consultancy; Amgen: Consultancy; Legend: Consultancy; Adaptive: Consultancy; Janssen: Consultancy; C4: Current equity holder in private company; OncoPep: Consultancy, Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties; BMS: Consultancy. Anderson: Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Millenium-Takeda: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Sanofi-Aventis: Membership on an entity's Board of Directors or advisory committees; Oncopep and C4 Therapeutics.: Other: Scientific Founder of Oncopep and C4 Therapeutics.; Celgene: Membership on an entity's Board of Directors or advisory committees.

Previous Abstract | Next Abstract >>
*signifies non-member of ASH