scholarly journals IL-15 Preconditioning Augments CAR T Cell Responses to Checkpoint Blockade for Improved Treatment of Solid Tumors

2020 ◽  
Vol 28 (11) ◽  
pp. 2379-2393
Author(s):  
Lauren Giuffrida ◽  
Kevin Sek ◽  
Melissa A. Henderson ◽  
Imran G. House ◽  
Junyun Lai ◽  
...  
2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A438-A438
Author(s):  
Mara Shainheit ◽  
Devin Champagne ◽  
Gabriella Santone ◽  
Syukri Shukor ◽  
Ece Bicak ◽  
...  

BackgroundATLASTM is a cell-based bioassay that utilizes a cancer patient‘s own monocyte-derived dendritic cells and CD4+ and CD8+ T cells to screen their mutanome and identify neoantigens that elicit robust anti-tumor T cell responses, as well as, deleterious InhibigensTM.1 GEN-009, a personalized vaccine comprised of 4–20 ATLAS-identified neoantigens combined with Hiltonol®, harnesses the power of neoantigen-specific T cells to treat individuals with solid tumors. The safety and efficacy of GEN-009 is being assessed in a phase 1/2a clinical trial (NCT03633110).MethodsA cohort of 15 adults with solid tumors were enrolled in the study. During the screening period, patients received standard of care PD-1-based immunotherapies appropriate for their tumor type. Subsequently, patients were immunized with GEN-009 with additional doses administered at 3, 6, 12, and 24 weeks. Peripheral blood mononuclear cells (PBMCs) were collected at baseline, pre-vaccination (D1), as well as 29, 50, 92, and 176 days post first dose. Vaccine-induced immunogenicity and persistence were assessed by quantifying neoantigen-specific T cell responses in ex vivo and in vitro stimulation dual-analyte fluorospot assays. Polyfunctionality of neoantigen-specific T cells was evaluated by intracellular cytokine staining. Additionally, potential correlations between the ATLAS-identified profile and vaccine-induced immunogenicity were assessed.ResultsGEN-009 augmented T cell responses in 100% of evaluated patients, attributable to vaccine and not checkpoint blockade. Furthermore, neoantigen-induced secretion of IFNγ and/or TNFα by PBMCs, CD4+, and CD8+ T cells was observed in all patients. Responses were primarily from polyfunctional TEM cells and detectable in both CD4+ and CD8+ T cell subsets. Some patients had evidence of epitope spreading. Unique response patterns were observed for each patient with no apparent relationship between tumor types and time to emergence, magnitude or persistence of response. Ex vivo vaccine-induced immune responses were observed as early as 1 month, and in some cases, persisted for 176 days. Clinical efficacy possibly attributable to GEN-009 was observed in several patients, but no correlation has yet been identified with neoantigen number or magnitude of immune response.ConclusionsATLAS empirically identifies stimulatory neoantigens using the patient‘s own immune cells. GEN-009, which is comprised of personalized, ATLAS-identified neoantigens, elicits early, long-lasting and polyfunctional neoantigen-specific CD4+ and CD8+ T cell responses in individuals with advanced cancer. Several patients achieved clinical responses that were possibly attributable to vaccine; efforts are underway to explore T cell correlates of protection. These data support that GEN-009, in combination with checkpoint blockade, represents a unique approach to treat solid tumors.AcknowledgementsWe are grateful to the patients and their families who consented to participate in the GEN-009-101 clinical trial.Trial RegistrationNCT03633110Ethics ApprovalThis study was approved by Western Institutional Review Board, approval number 1-1078861-1. All subjects contributing samples provided signed individual informed consent.ReferenceDeVault V, Starobinets H, Adhikari S, Singh S, Rinaldi S, Classon B, Flechtner J, Lam H. Inhibigens, personal neoantigens that drive suppressive T cell responses, abrogate protection of therapeutic anti-tumor vaccines. J. Immunol 2020; 204(1 Supplement):91.15.


JCI Insight ◽  
2016 ◽  
Vol 1 (19) ◽  
Author(s):  
Yogindra Vedvyas ◽  
Enda Shevlin ◽  
Marjan Zaman ◽  
Irene M. Min ◽  
Alejandro Amor-Coarasa ◽  
...  

2020 ◽  
Vol 28 (7) ◽  
pp. 1563-1566
Author(s):  
Amanda X.Y. Chen ◽  
Imran G. House ◽  
Paul A. Beavis ◽  
Phillip K. Darcy

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3694-3694
Author(s):  
Anne W. J. Martens ◽  
Felix M Wensveen ◽  
Ramon Arens ◽  
Eric Eldering ◽  
Gerritje J. W. van der Windt ◽  
...  

Abstract Background - Tumor immunosuppression is a major cause for treatment failure and disease relapse, both in solid tumors and in leukemia. New T cell mediated therapeutic modalities (such as CAR T cells, bispecific antibodies and immune checkpoint blockade) can be highly effective, but depend on autologous T cell functionality. In chronic lymphocytic leukemia (CLL), ex vivo patient studies have shown that T cells are skewed toward a terminally differentiated, dysfunctional phenotype, which may contribute to the disappointing results of T cell therapies in this disease. Recently, it has been shown that for CAR T cell efficacy in CLL, the presence of memory subsets both prior to CAR T cell generation and in the infusion product, is instrumental for CAR T cell persistence and complete responses (Fraietta et al., 2018). However, it is currently unknown how the presence of CLL impacts T cell skewing and whether it affects acute antigen-specific immune responses in vivo. As dynamic immune studies are limited in the human setting, we investigated whether acute antigen-mediated T cell responses are affected by CLL using the TCL1 adoptive transfer mouse model. Earlier T cell studies in this model revealed similar changes as seen in the human disease, both with respect to gene expression profiles as well as phenotypic and functional characteristics. Methods - C57BL/6J (CD45.2+) mice were injected i.p. with 20x106 TCL1 transgenic splenocytes (CD45.2+) or PBS. During development of CLL mice were monitored for T cell subset differentiation. When CLL mice reached over 70% CLL in blood, 50.000 OT-I cells (CD45.1+), which specifically recognize ovalbumin) were adoptively transferred (i.v.) into all mice, directly followed by infection with 100.000 PFU mCMV-OVA. This model allowed us to study the endogenous (CD45.2+) T cell compartment during CLL development, as well as the acute response of naïve, antigen-specific (CD45.1+) T cells that had not been influenced by CLL prior to antigen exposure. Seven days after infection the mice were sacrificed and splenocytes were analyzed by flow cytometry either directly or after re-stimulation with OVA-peptide to assess cytokine production. Results - Using this adoptive transfer model of CLL, we found that CLL mice showed a decrease in naïve CD8 T cells, an increase of antigen-experienced cells, and a reversed CD4/CD8 ratio compared to control mice, as reported earlier (McClanahan et al., 2015). At day 7 after injection of naïve OT-I cells and infection with mCMV-OVA, percentages of OT-I cells were similar in CLL and control mice, and all OT-I cells showed an effector phenotype (CD44+CD62L-). However, in this effector OT-I pool CLL mice showed an increased frequency of KLRG1+CD127- short-lived effector cells (SLEC), while KLRG1-CD127+ memory precursor effector cells (MPEC) were decreased compared to control mice. This was associated with an enhanced expression of the effector-associated transcription factor T-bet, and reduced expression of the memory-associated transcription factor Bcl-6 in total OT-I cells and within the OT-I SLEC and MPEC populations of CLL mice. Since poor responses to CAR T cell therapy are associated with effector phenotypes and higher rates of glycolysis (Fraietta et al., 2018), and Bcl-6 directly represses glycolysis (Oestreich et al., 2014), we analyzed glucose uptake by OT-I cells. In line with reduced expression of Bcl-6, we found increased levels of glucose uptake in CLL derived OT-I cells. Despite the skewing towards a more short-lived effector phenotype of the CLL derived OT-I cells, in vitro re-stimulation with OVA peptide resulted in decreased production of IFN-γ and decreased degranulation as measured by CD107a (LAMP-1) expression compared to control derived OT-I cells. Conclusion - Our findings show that in this mouse model CLL development skews the T cell compartment towards more antigen-experienced cells. In addition to this, our results suggest that there is a CLL-antigen independent effect on acute antigen-specific immune responses. This CLL mediated effect drives T cells towards an effector phenotype, that is functionally impaired. This study provides clues for better understanding T cell responses in CLL, and may lead to new strategies to improve T cell mediated therapies in CLL, which are currently being exploited. Disclosures Eldering: Celgene: Research Funding. Kater:Acerta: Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche/Genentech: Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Research Funding.


Cancers ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 743
Author(s):  
Aleksei Titov ◽  
Ekaterina Zmievskaya ◽  
Irina Ganeeva ◽  
Aygul Valiullina ◽  
Alexey Petukhov ◽  
...  

Adoptive cell immunotherapy (ACT) is a vibrant field of cancer treatment that began progressive development in the 1980s. One of the most prominent and promising examples is chimeric antigen receptor (CAR) T-cell immunotherapy for the treatment of B-cell hematologic malignancies. Despite success in the treatment of B-cell lymphomas and leukemia, CAR T-cell therapy remains mostly ineffective for solid tumors. This is due to several reasons, such as the heterogeneity of the cellular composition in solid tumors, the need for directed migration and penetration of CAR T-cells against the pressure gradient in the tumor stroma, and the immunosuppressive microenvironment. To substantially improve the clinical efficacy of ACT against solid tumors, researchers might need to look closer into recent developments in the other branches of adoptive immunotherapy, both traditional and innovative. In this review, we describe the variety of adoptive cell therapies beyond CAR T-cell technology, i.e., exploitation of alternative cell sources with a high therapeutic potential against solid tumors (e.g., CAR M-cells) or aiming to be universal allogeneic (e.g., CAR NK-cells, γδ T-cells), tumor-infiltrating lymphocytes (TILs), and transgenic T-cell receptor (TCR) T-cell immunotherapies. In addition, we discuss the strategies for selection and validation of neoantigens to achieve efficiency and safety. We provide an overview of non-conventional TCRs and CARs, and address the problem of mispairing between the cognate and transgenic TCRs. Finally, we summarize existing and emerging approaches for manufacturing of the therapeutic cell products in traditional, semi-automated and fully automated Point-of-Care (PoC) systems.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A444-A444
Author(s):  
Cathy Eng ◽  
Joaquina Baranda ◽  
Matthew Taylor ◽  
Michael Gordon ◽  
Ursula Matulonis ◽  
...  

BackgroundSQZ-PBMC-HPV is a therapeutic cancer vaccine created with Cell Squeeze®, a proprietary cell-engineering system. SQZ-PBMC-HPV is a novel cancer vaccine generated from peripheral blood mononuclear cells (PBMC) squeezed with HPV16 E6 and E7 antigens, resulting in delivery into the cytosol. The resulting antigen presenting cells (APCs) provide enhanced antigen presentation on MHC-I to potentially elicit robust, antigen-specific CD8+ T cell responses. Importantly, SQZ-PBMC-HPV are neither genetically modified nor immune effector cells.Studies in MHC-I knockout mice demonstrated that activation of antigen specific CD8+ tumor infiltrating lymphocytes (TILs) was a direct effect of cytosolic antigen delivery to PBMCs. In the murine TC-1 tumor model, tumor regression correlated with an influx of HPV16-specific CD8+ TILs. In vitro studies with human volunteer PBMCs demonstrated that each subset is capable of inducing CD8+ T cell responses. The Phase 1 study includes a significant biomarker program to investigate whether pharmacodynamic effects observed in non-clinical studies correlate with potential clinical benefit. Immunogenic and pharmacodynamic endpoints include Elispot assays to measure frequency of interferon gamma secreting cells, as well as quantification and characterization of TILs and tumor microenvironment. In addition, various cytokine responses and circulating cell-free HPV16 DNA levels in plasma are measured.MethodsSQZ-PBMC-HPV-101 (NCT04084951) is open for enrollment to HLA A*02+ patients with HPV16+ recurrent, locally advanced or metastatic solid tumors and includes escalation cohorts for monotherapy and in combination with atezolizumab. After initial demonstration of safety, the study assesses dose effect by testing different cell dose levels, the effect of prolonged antigen priming in Cycle 1 [APC administration on Day 1 only compared to Days 1 and 2 (double priming)] and the impact of treatment duration to identify the optimal dose regimen. The cycle length is 3 weeks, and patients will receive SQZ-PBMC-HPV for up to 1 year or until available autologous drug product is exhausted. Atezolizumab will be administered for up to 1 year. Eligible patients including but not limited to anal, cervical and head and neck tumors will undergo a single leukapheresis at the study site. The manufacturing process includes a maturation step and takes less than 24 hours. The vein-to-vein time for the 1st administration is approximately one week. Patients must have a lesion that can be biopsied with acceptable clinical risk and agree to have a fresh biopsy at Screening and on study. A Study Safety Committee is in place. No formal statistical hypothesis testing will be performed.ResultsN/AConclusionsN/ATrial RegistrationNCT04084951Ethics ApprovalThe study is registered on clinicaltrials.gov was approved by the Ethics Board of all institution listed as recruiting.


Author(s):  
Öykü Umut ◽  
Adrian Gottschlich ◽  
Stefan Endres ◽  
Sebastian Kobold

SummaryChimeric antigen receptor (CAR) T cell therapy has been established in the treatment of hematological malignancies. However, in solid tumors its efficacy remains limited. The aim of this article is to give an overview of the field of cell therapy itself, to introduce the underlying concepts of CAR T cell-based treatment approaches and to address its limitations in advancing the treatment for solid malignancies.


Author(s):  
Pooria Safarzadeh Kozani ◽  
Pouya Safarzadeh Kozani ◽  
Fatemeh Rahbarizadeh

Cancers ◽  
2020 ◽  
Vol 12 (8) ◽  
pp. 2087
Author(s):  
Yuna Jo ◽  
Laraib Amir Ali ◽  
Ju A Shim ◽  
Byung Ha Lee ◽  
Changwan Hong

Novel engineered T cells containing chimeric antigen receptors (CAR-T cells) that combine the benefits of antigen recognition and T cell response have been developed, and their effect in the anti-tumor immunotherapy of patients with relapsed/refractory leukemia has been dramatic. Thus, CAR-T cell immunotherapy is rapidly emerging as a new therapy. However, it has limitations that prevent consistency in therapeutic effects in solid tumors, which accounts for over 90% of all cancer patients. Here, we review the literature regarding various obstacles to CAR-T cell immunotherapy for solid tumors, including those that cause CAR-T cell dysfunction in the immunosuppressive tumor microenvironment, such as reactive oxygen species, pH, O2, immunosuppressive cells, cytokines, and metabolites, as well as those that impair cell trafficking into the tumor microenvironment. Next-generation CAR-T cell therapy is currently undergoing clinical trials to overcome these challenges. Therefore, novel approaches to address the challenges faced by CAR-T cell immunotherapy in solid tumors are also discussed here.


Sign in / Sign up

Export Citation Format

Share Document