Experimental treatment of colorectal cancer in mice with human T cells electroporated with NKG2D RNA CAR

Immunotherapy ◽  
2020 ◽  
Author(s):  
Zhendong Li ◽  
Zhixia Chi ◽  
Wei-Xia Ang ◽  
Can Chen ◽  
Johan CK Tay ◽  
...  

Aim: Peritoneal metastasis is often present in end-stage neoplastic diseases, including recurrent colorectal cancer and is associated with decreased overall survival. Novel methods are needed. Materials & methods: We constructed first-, second- and third-generation chimeric antigen receptors (CARs) specific for NKG2D ligands and modified human T cells with mRNA electroporation. Results: NKG2D CAR expression was detectable for at least 6 days postelectroporation and mediated efficient cytotoxicity against NKG2DL+ tumor cells, but not NKG2DL-cells. Multiple infusions of the first-generation CAR-T cells into immunodeficient mice bearing established peritoneal colorectal xenografts led to significantly reduced tumor burden. Conclusion: mRNA CAR is an economical way to test new CARs and potentiates controlling on-target/off-tumor toxicity and cytokine storms. The use of NKG2D RNA CARs to treat colorectal peritoneal metastasis warrants further investigation.

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A717-A717
Author(s):  
Abigail Overacre-Delgoffe ◽  
Anthony Cillo ◽  
Hannah Bumgarner ◽  
Ansen Burr ◽  
Justin Tometich ◽  
...  

BackgroundColorectal cancer remains one of the most common and deadliest cancers worldwide and effective therapies are lacking. While immunotherapy has revolutionized treatment for many cancers, the overwhelming majority of colorectal cancer patients are non-responsive and the 5-year survival rate for advanced disease is <20%. Immunotherapeutic response has been associated with select members of the microbiome in melanoma; however, the potential benefit in colorectal cancer and the underlying mechanisms remain unclear. We sought to determine how specific members of the intestinal microbiome affect anti-tumor immunity in colorectal cancer (CRC) in hopes of discovering novel treatments and revealing potential hurdles to current therapeutic response in CRC patients.MethodsWe utilized a carcinogen-induced mouse model of CRC and colonized half of the tumor-bearing mice with Helicobacter hepaticus (Hhep) 7 weeks post AOM. Tumor number was assessed 12 weeks post AOM. We isolated lymphocytes from the lamina propria, colonic epithelium, mesenteric lymph nodes, and tumor(s) to track the spatial and transcriptional Hhep-specific and endogenous immune responses during tumor progression through 5’ single cell RNAseq, flow cytometry, and immunofluorescence. In addition, we utilized 16S sequencing and FISH to track Hhep colonization, location within the colon, and its impact on the surrounding microbiome.ResultsWe have found that rational modification of the microbiome of colon tumor-bearing mice through addition of a single bacteria, Hhep, led to tumor control or clearance and a significant survival advantage. Colonization led to the expansion of the lymphatic network and development of numerous peri- or intra-tumoral tertiary lymphoid structures (TLS) composed of Hhep-specific CD4 T follicular helper cells (TFH) as well as the bacteria itself. This led to an overall ‘heating’ of the tumor, wherein we saw an increase of CD4 T cell infiltration to the tumor core as well as an increase in CD103+ type 1 DC (cDC1) recruitment through increased chemokines such as CCL5 and XCL1. Hhep-specific TFH were both necessary and sufficient to drive TLS formation, increased immune invasion, and anti-tumor immunity.ConclusionsWe have shown that addition of a single bacteria, Hhep, leads to a reduction in CRC tumor burden or clearance through lymphatic expansion, TLS formation, and remodeling of the tumor microenvironment, and that Hhep-specific T cells are required for tumor control. These studies suggest that rational modification of the microbiome and microbiome-specific T cells can positively impact anti-tumor immunity and may represent a unique immunotherapeutic target to turn resistant tumors into responsive tumors.


2012 ◽  
Vol 24 (4) ◽  
pp. 476-481 ◽  
Author(s):  
Sian M Henson ◽  
Natalie E Riddell ◽  
Arne N Akbar
Keyword(s):  
T Cells ◽  

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A880-A880
Author(s):  
Abigail Overacre-Delgoffe ◽  
Hannah Bumgarner ◽  
Anthony Cillo ◽  
Ansen Burr ◽  
Justin Tometich ◽  
...  

BackgroundColorectal cancer (CRC) is one of the most common and deadly cancers in the US, and the survival rate for advanced cases is poor. While immunotherapy has revolutionized cancer treatment, CRC remains largely unresponsive, with only ~6% of patients responding to anti-PD1. Specific microbiome signatures are associated with anti-PD1 response in melanoma patients; however, the underlying mechanism remains unclear. While the microbiome in cancer patients has been extensively studied, the endogenous immune response to these microbes and the subsequent effects on cancer immunity remain unstudied. Most microbes reside within the gut, and bacteria that adhere to the intestinal epithelium can stimulate bacteria-specific immune responses. Therefore, we hypothesized that the microbiome, especially adherent, immunogenic bacteria, may support anti-tumor immunity through activation of local microbiota-specific T cells.MethodsUsing a carcinogen-induced mouse model of CRC, we sought to determine the impact of microbiome modulation on the anti-tumor immune response. We colonized tumor-bearing mice with Helicobacter hepaticus (Hhep) and assessed tumor burden, survival, and immune infiltration. Lymphocytes were isolated from the tumor and surrounding tissue when tumors were terminal (12 weeks). We utilized TCR transgenic mice and MHC class II tetramers to track the spatial and transcriptional Hhep-specific T cell response through 5’ single cell RNAseq, flow cytometry, and spectral immunofluorescence.ResultsHhep colonization in tumor-bearing mice led to decreased tumor burden and significantly improved survival. Interestingly, colonization induced activation of Hhep-specific T follicular helper cells (TFHs) that supported formation of mature peri- or intra-tumoral tertiary lymphoid structures (TLS). The presence of TLS led to increased infiltration of cytotoxic lymphocytes (T and NK cells) within the tumor core. Surprisingly, the anti-tumor response was dependent on CD4+ T and B cells but not CD8+ T cells. Using TFH KO mice, we found that Hhep-specific CD4+ T cells were both necessary and sufficient to drive TLS maturation and anti-tumor immunity.ConclusionsHere, we demonstrate that addition of a single bacterial species after tumor formation leads to a reduction in CRC tumor burden and increased survival through TLS maturation. This microbiome-dependent remodeling of the tumor microenvironment is driven by Hhep-specific TFH cells that are both necessary and sufficient for tumor control, demonstrating for the first time that microbiota-specific T cells contribute to anti-tumor immunity. Overall, these findings suggest that microbiome modulation and the subsequent microbiota-specific CD4+ T cell response may represent a new variety of immunotherapies for cancers that remain resistant to checkpoint blockade.


2020 ◽  
Vol 4 (21) ◽  
pp. 5343-5356
Author(s):  
Laura R. E. Becher ◽  
Wendy K. Nevala ◽  
Shari Lee Sutor ◽  
Megan Abergel ◽  
Michele M. Hoffmann ◽  
...  

Abstract Human cytomegalovirus (HCMV) induces long-lasting T-cell immune responses that control but do not clear infection. Typical responses involve private T-cell clones, expressing T-cell antigen receptors (TCRs) unique to a person, and public T-cell clones with identical TCRs active in different people. Here, we report the development of a pretherapeutic immunostimulation modality against HCMV for human T cells, CD3 copotentiation, and the clonal analysis of its effects in recall assays at single-cell resolution. CD3 copotentiation of human T cells required identification of an intrinsically inert anti-CD3 Fab fragment that conditionally augmented signaling only when TCR was coengaged with antigen. When applied in recall assays, CD3 copotentiation enhanced the expansion of both public and private T-cell clones responding to autologous HLA-A2(+) antigen-presenting cells and immunodominant NLVPMVATV (NLV) peptide from HCMV pp65 protein. Interestingly, public vs private TCR expression was associated with distinct clonal expansion signatures in response to recall stimulus. This implied that besides possible differences in their generation and selection in an immune response, public and private T cells may respond differently to pharmacoimmunomodulation. Furthermore, a third clonal expansion profile was observed upon CD3 copotentiation of T-cell clones from HLA-A2(−) donors and 1 HLA-A2(+) presumed-uninfected donor, where NLV was of low intrinsic potency. We conclude that human T-cell copotentiation can increase the expansion of different classes of T-cell clones responding to recall antigens of different strengths, and this may be exploitable for therapeutic development against chronic, persistent infections such as HCMV.


2017 ◽  
Vol 35 (4_suppl) ◽  
pp. 605-605 ◽  
Author(s):  
Julie G. Grossman ◽  
Timothy M. Nywening ◽  
Brian Belt ◽  
Michael Ahlers ◽  
William G. Hawkins ◽  
...  

605 Background: Colorectal cancer (CRC) is the most common gastrointestinal malignancy. 60% of CRC patients are diagnosed with metastatic CRC (mCRC) and the 5-year survival is < 20%. CCR2+ inflammatory monocytes (IM) are recruited from the bone marrow to the tumor microenvironment by the CCL2/CCR2 chemokine axis. At the tumor, they become tumor associated macrophages (TAM) and play a crucial role in promoting tumor progression, metastasis, and chemoresistance. While the importance of IM has been shown in other malignancies, little is known about their role in mCRC. Methods: Flow cytometry was performed on human and murine PBMCs, adjacent normal tissue, and tumors. Qualitative RT-PCR, ELISA, and confocal microscopy were performed for CCL2 expression. T-cell suppression assays were performed using CD14+ IM isolated from patient PBMCs and liver metastasis (LM). A murine model of CRC LM was created by hemispleen injection of luciferase-labelled MC38 CRC cells. Mice were treated with a CCR2 inhibitor and/or FOLFOX. Results: Prior to liver resection, mCRC patients have a higher percentage of IM in the peripheral blood compared to healthy donors (p < 0.0001), and on multivariate analysis elevated monocytes were prognostic of poor survival. We also found higher levels of CCL2 in the serum of mCRC patients (p < 0.01). Additionally, there was increased expression of CCL2 in LM compared to uninvolved tissue (p < 0.01), with the production of CCL2 localized to mCRC cells. FACS analysis showed CCR2+ TAM were elevated in LM compared to adjacent normal liver (p < 0.05) with a paucity of effector T-cells. CD14+ TAMs isolated from mCRC inhibited T-cell proliferation, illustrating the immune suppressive phenotype of these cells. In a murine model of CRC LM, treating mice with a CCR2 inhibitor alone or in combination with FOLFOX chemotherapy resulted in decreased tumor burden. FACS analysis of treated tumors showed increased effector T-cells in mice treated with CCR2 inhibitor. Conclusions: IM are involved in the progression of mCRC. Targeting these immunosuppressive cells with a CCR2 inhibitor decreases tumor burden in a murine model. This represents a potential novel treatment for mCRC.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 192-192
Author(s):  
Adrienne H. Long ◽  
Rimas J. Orentas ◽  
Crystal L. Mackall

Abstract Introduction Chimeric antigen receptors (CARs) provide a promising new approach for the adoptive immunotherapy of cancer. Though impressive antitumor activity has been observed with some CAR T cells, other CAR T cells demonstrate poor antitumor efficacy in vivo despite high cytolytic capacity in vitro due to poor expansion and persistence. Whether exhaustion of CAR T cells mirrors exhaustion that occurs naturally in chronically stimulated human T cells has not yet been studied. Here, we report that expression of select CD28 containing CARs in normal human T cells rapidly induces an exhausted state characterized by high PD-1 expression, poor persistence and poor antitumor efficacy, whereas other CARs do not induce this phenotype. Results Human T cells were expanded with anti-CD3/CD28 beads, and then transduced with a second-generation (CD28-CD3ζ) disialoganglioside 2 (GD2) specific CAR or a second-generation (CD28-CD3ζ) CD19 specific CAR. By day 7 of in vitro expansion, GD2 CAR T cells developed a metabolism more highly dependent on glycolysis compared to CD19 CAR T cells or untransduced controls. Neither CAR population was exposed to antigen during this expansion period. Using a Seahorse Extracellular Flux Analyzer, the ratio of glycolysis to oxidative phosphorylation rates (ECAR:OCR ratio) of GD2 CAR T cells was found to be double that of CD19 CAR T cells or controls on day 7. The highly glycolytic metabolism of GD2 CAR T cells was associated with an exhausted phenotype. GD2 CAR T cells expressed higher levels of PD-1, TIM-3 and LAG-3, and transcription repressor BLIMP-1, compared to CD19 CAR T cells or untransduced controls. Additionally, GD2 CAR T cells were poor cytokine producers, generating <10x lower levels of IL2, TNFα and IFNγ than CD19 CAR T cells upon in vitro co-incubation with a GD2+CD19+ osteosarcoma line (143B-CD19), despite maintaining comparable in vitro cytolytic ability. GD2 CAR T cells showed poor in vitro expansion and increased rates of apoptosis compared to controls. GD2 CAR T cells also did not persist and did not mediate antitumor effects against GD2+CD19+ tumors in a murine xenograft model in vivo, whereas CD19 CAR T cells completely eradicated CD19+ tumors and persisted in both the spleen and tumor compartments. To rule out the possibility that diminished cytokine production and in vivo efficacy was related to antigen specific effects, T cells were co-transduced with both the GD2 and CD19 CARs. Though single-transduced CD19 CAR T cells show no signs of an altered metabolism or exhaustion and have strong antitumor efficacy, CD19 CAR T cells co-transduced with the GD2 CAR demonstrate an exhausted phenotype and diminished antitumor efficacy similar to that of single-transduced GD2 CAR T cells. Thus, expression of the GD2 CAR confers a dominant exhausted phenotype in T cells, and prevents otherwise efficacious CARs from mediating strong antitumor effects. We hypothesized that chronic signaling of CD3ζ and CD28 via the GD2 CAR results in exhaustion. Interestingly, however, we did not identify GD2 expression in the culture system. Point mutations in the CAR antigen-binding site, though abrogating GD2 binding, did not prevent the development of exhaustion. Thus, we postulate that constitutive receptor signaling may occur via interactions between the framework regions of the CAR receptors. Importantly however, substitution of 4-1BB for the CD28 domain in the GD2 CAR substantially diminished PD-1 expression, one of the hallmark features of exhausted T cells. Conclusions We report that expression of a CD28 containing GD2 CAR induces both an altered metabolism and an exhausted state in human T cells, resulting in poor in vivo persistence and antitumor efficacy. We hypothesize that tonic signaling through the GD2 CAR induces this phenotype and have identified the CD28 domain as an important component contributing to this phenotype. Rapid induction of exhaustion mediated via a synthetic receptor provides a novel model system to identify mechanistic factors required for this phenotype in human T cells. Work is currently underway to molecularly define the basis for the exhaustion of GD2 CAR T cells and to probe a potential role for altered T cell metabolism as a contributor to T cell exhaustion in human T cells. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 1 (2) ◽  
pp. 7 ◽  
Author(s):  
Ana C. Parente-Pereira ◽  
Scott Wilkie ◽  
Sjoukje J.C. Van der Stegen ◽  
David M. Davies ◽  
John Maher

Blood ◽  
2010 ◽  
Vol 115 (25) ◽  
pp. 5154-5163 ◽  
Author(s):  
Ralf-Holger Voss ◽  
Simone Thomas ◽  
Christina Pfirschke ◽  
Beate Hauptrock ◽  
Sebastian Klobuch ◽  
...  

Abstract Transfer of tumor antigen–specific T-cell receptors (TCRs) into human T cells aims at redirecting their cytotoxicity toward tumors. Efficacy and safety may be affected by pairing of natural and introduced TCRα/β chains potentially leading to autoimmunity. We hypothesized that a novel single-chain (sc)TCR framework relying on the coexpression of the TCRα constant α (Cα) domain would prevent undesired pairing while preserving structural and functional similarity to a fully assembled double-chain (dc)TCR/CD3 complex. We confirmed this hypothesis for a murine p53-specific scTCR. Substantial effector function was observed only in the presence of a murine Cα domain preceded by a TCRα signal peptide for shuttling to the cell membrane. The generalization to a human gp100-specific TCR required the murinization of both C domains. Structural and functional T-cell avidities of an accessory disulfide-linked scTCR gp100/Cα were higher than those of a dcTCR. Antigen-dependent phosphorylation of the proximal effector ζ-chain–associated protein kinase 70 at tyrosine 319 was not impaired, reflecting its molecular integrity in signaling. In melanoma-engrafted nonobese diabetic/severe combined immunodeficient mice, adoptive transfer of scTCR gp100/Cα transduced T cells conferred superior delay in tumor growth among primary and long-term secondary tumor challenges. We conclude that the novel scTCR constitutes a reliable means to immunotherapeutically target hematologic malignancies.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2902-2902 ◽  
Author(s):  
Omkar Uday Kawalekar ◽  
Avery D. Posey ◽  
Joseph Fraietta ◽  
Jihyun Lee ◽  
John Scholler ◽  
...  

Abstract Background Chimeric Antigen Receptors (CARs) have shown great promise in the field of targeted adoptive immunotherapy against cancer. These receptors are specific for tumor antigens and have the binding properties of monoclonal antibodies with signaling molecules of T cells. When expressed on T cells, these receptors help the cells home to tumor targets and perform their cytotoxic functions. CARs containing the 4-1BB signaling domain have been used against B-cell chronic lymphocytic leukemia and have shown the most clinical success in terms of tumor targeting and persistence in patients upon engraftment. In contrast, their CD28-containing CAR counterpart failed to show comparable persistence in patients. Despite extensive clinical use, the detailed molecular mechanisms involved in the activation of CAR-grafted T cells remain elusive. To address this, we hypothesize that CARs take advantage of the endogenous T cell receptor (TCR) signaling pathways in a manner unique to their analogous intracellular domains. Methods By electroporation of CAR encoding in vitro transcribed RNA into primary human T cells, we achieved >90% CAR-positive T cell population. We expressed different CARs constructs, all specific for a widely expressed tumor antigen - mesothelin. Keeping the scFv region constant to SS1 that is specific for mesothelin, we varied the intracellular signaling domains (ICDs) ranging from first generation CARs (containing only the CD3z ICD) to the second generation CARs (CD28-CD3z or 41BB-CD3z ICDs) Upon verifying CAR expression by flow cytometry, these T cells were stimulated with mesothelin antigen to analyze differences in signaling between the different CAR groups. Results Here we report that CARs with CD28 show stronger activation of T cells when compared to CARs with 4-1BB or CD3z alone. Stimulation of different CAR constructs revealed that the antigen-specific activation threshold for CAR-T cells is greatly reduced when the CD28 endodomain is included in the CAR architecture. This activation state, measured by the activation of proximal signaling proteins, as well as the MAPK and Akt signaling pathways continues to increase and persist for longer time durations in T cells with the CD28-containing CAR construct. Co-immunoprecipitation studies reveal direct interaction of CARs with pZAP70 and TRAF proteins, but not other known signaling molecules of the TCR complex. T cells with CARs containing CD28 intracellular domain showed a high and sustained level of calcium flux in comparison to T cells with the 4-1BB containing CARs. Experiments to determine the molecular signatures of CAR-grafted T cells stimulated with cognate antigen for longer time durations are currently underway. Taken together, these studies have significant impact on the future design of CARs and adoptive immunotherapy. Disclosures: Kawalekar: Novartis: Research Funding. Posey:Novartis: Research Funding. Fraietta:Novartis: Research Funding. Lee:Novartis: Research Funding. Zhao:Novartis: Research Funding. June:Novartis: Research Funding.


PLoS ONE ◽  
2015 ◽  
Vol 10 (6) ◽  
pp. e0130518 ◽  
Author(s):  
Maud Condomines ◽  
Jon Arnason ◽  
Reuben Benjamin ◽  
Gertrude Gunset ◽  
Jason Plotkin ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document