Journal Articles

Regulatory T cells define affinity thresholds for CD8+ T cell tumor infiltration

Mohsen et al. showed that a low-affinity LCMV p33-derived A4Y peptide linked to a bacteriophage Qβ virus-like particle induced a robust and specific T cell response, but showed limited cross-reactivity to p33 and failed to protect against B16F10p33 tumor growth. Qβ-A4Y combined with Treg-depleting anti-CD25 increased CD8+ T cell infiltration into tumors and migration away from blood vessels. Treg depletion enhanced the antitumor activity of Qβ-A4Y, promoted T cell-mediated tumor-free survival, enhanced T cell cross-reactivity, and increased the effectiveness of a vaccine targeting multiple naturally low-affinity tumor-associated antigens.

Contributed by Shishir Pant

Mohsen et al. showed that a low-affinity LCMV p33-derived A4Y peptide linked to a bacteriophage Qβ virus-like particle induced a robust and specific T cell response, but showed limited cross-reactivity to p33 and failed to protect against B16F10p33 tumor growth. Qβ-A4Y combined with Treg-depleting anti-CD25 increased CD8+ T cell infiltration into tumors and migration away from blood vessels. Treg depletion enhanced the antitumor activity of Qβ-A4Y, promoted T cell-mediated tumor-free survival, enhanced T cell cross-reactivity, and increased the effectiveness of a vaccine targeting multiple naturally low-affinity tumor-associated antigens.

Contributed by Shishir Pant

ABSTRACT: TCR repertoires against tumors lack high-affinity TCRs and are further suppressed by Tregs. We hypothesized that Treg depletion enhances the antitumor efficacy of low-affinity T cells. Using the weak agonistic peptide A4Y derived from LCMV glycoprotein peptide p33 as a model antigen and VLPs as a vaccine platform, we tested this approach. In a separate low-affinity model, we targeted B16F10 melanoma with our multi-target vaccine. Results revealed limited in vivo lytic cross-reactivity between A4Y and p33 peptides, and the A4Y-vaccine alone failed to inhibit B16F10p33 tumor progression. However, combining A4Y-vaccine with Treg depletion triggered a robust immune response, characterized by increased CD8+ T cell infiltration, enhanced T cell functionality, and tumor-free survival. Infiltrating T cells also exhibited closer spatial proximity and heightened migration from blood vessels. Similarly, combining low-affinity vaccine with Treg depletion enhanced antitumor responses. These findings highlight the potential of Treg depletion to advance vaccination strategies targeting TAAs with low-affinity T cells.

Author Info: (1) Department of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland. mona.mohsen@unibe.ch. Department for BioMedical Research, University of Bern, Ber

Author Info: (1) Department of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland. mona.mohsen@unibe.ch. Department for BioMedical Research, University of Bern, Bern, Switzerland. mona.mohsen@unibe.ch. DeepVax GmbH, 8487 RŠmismŸhle, ZŸrich, Switzerland. mona.mohsen@unibe.ch. (2) Department of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland. Department for BioMedical Research, University of Bern, Bern, Switzerland. Graduate School for Cellular and Biomedical Sciences (GCB), Bern, Switzerland. (3) Department for BioMedical Research, University of Bern, Bern, Switzerland. (4) Department of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland. Department for BioMedical Research, University of Bern, Bern, Switzerland. (5) Department of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland. Department for BioMedical Research, University of Bern, Bern, Switzerland. (6) Department of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland. Department for BioMedical Research, University of Bern, Bern, Switzerland. Graduate School for Cellular and Biomedical Sciences (GCB), Bern, Switzerland. (7) Department of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland. Department for BioMedical Research, University of Bern, Bern, Switzerland. Graduate School for Cellular and Biomedical Sciences (GCB), Bern, Switzerland. (8) Department for BioMedical Research, University of Bern, Bern, Switzerland. DeepVax GmbH, 8487 RŠmismŸhle, ZŸrich, Switzerland. Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland. (9) COMPATH, Institute of Animal Pathology, University of Bern, Bern, Switzerland. (10) Department of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland. Department for BioMedical Research, University of Bern, Bern, Switzerland. Nuffield Department of Medicine, The Henry Welcome Building for Molecular Physiology, The Jenner Institute, University of Oxford, Oxford, UK.

Targeting the CD40 costimulatory receptor to improve virotherapy efficacy in diffuse midline gliomas

Labiano et al. showed that in mice with diffuse midline glioma (DMG), intratumoral co-administration of the Delta-24-RGD oncolytic virus and agonist CD40 antibodies extended survival and induced complete responses in 40% of mice, with protection from rechallenge that was dependent on local and resident immune memory. No signs of toxicity were observed. Mechanistically, treatment induced TME remodeling towards a pro-inflammatory landscape. Macrophage and microglia mediated recruitment of mature, cross-presenting cDC1s that supported the accumulation of activated and proliferating CD4+ and CD8+ TILs in tumors.

Contributed by Lauren Hitchings

Labiano et al. showed that in mice with diffuse midline glioma (DMG), intratumoral co-administration of the Delta-24-RGD oncolytic virus and agonist CD40 antibodies extended survival and induced complete responses in 40% of mice, with protection from rechallenge that was dependent on local and resident immune memory. No signs of toxicity were observed. Mechanistically, treatment induced TME remodeling towards a pro-inflammatory landscape. Macrophage and microglia mediated recruitment of mature, cross-presenting cDC1s that supported the accumulation of activated and proliferating CD4+ and CD8+ TILs in tumors.

Contributed by Lauren Hitchings

ABSTRACT: Diffuse midline glioma (DMG) is a devastating pediatric brain tumor. The oncolytic adenovirus Delta-24-RGD has shown promising efficacy and safety in DMG patients but is not yet curative. Thus, we hypothesized that activating dendritic cells (DCs) through the CD40 costimulatory receptor could increase antigen presentation and enhance the anti-tumor effect of the virus, resulting in long-term responses. This study shows that the intratumoral co-administration of Delta-24-RGD and a CD40 agonistic antibody is well tolerated and induces long-term anti-tumor immunity, including complete responses (up to 40%) in DMG preclinical models. Mechanistic studies revealed that this therapy increased tumor-proliferating T lymphocytes and proinflammatory myeloid cells, including mature DCs with superior tumor antigen uptake capacity. Moreover, the lack of cross-presenting DCs and the prevention of DC recruitment into the tumor abolish the Delta-24-RGD+anti-CD40 anti-DMG effect. This approach shows potential for combining virotherapy with activating antigen-presenting cells in these challenging tumors.

Author Info: (1) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pampl

Author Info: (1) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. Electronic address: slalminana@unav.es. (2) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (3) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (4) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (5) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (6) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (7) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (8) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (9) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (10) Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA. (11) Bioinformatics Platform, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain. (12) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (13) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (14) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (15) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (16) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (17) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (18) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (19) Jack Martin Fund Division of Pediatric Hematology-oncology, Mount Sinai, New York, NY, USA. (20) Dpt. Of NeuroOncology, UT MD Anderson Cancer Center, Houston, TX, USA. (21) Dpt. Of NeuroOncology, UT MD Anderson Cancer Center, Houston, TX, USA. (22) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (23) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. (24) Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain; Program in Solid Tumors, Center for the Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain. Electronic address: mmalonso@unav.es.

A CXCR4 partial agonist improves immunotherapy by targeting immunosuppressive neutrophils and cancer-driven granulopoiesis

Qian and Ma et al. fused TFF2, a partial CXCR4 agonist, with murine serum albumin (MSA) to develop TFF2–MSA peptides that restore anti-PD-1 sensitivity in syngeneic gastric cancer (GC) tumor models. TFF2-MSA distinctly modulated CXCR4 signaling, reducing bone marrow granulopoiesis compared to full agonists. TFF2–MSA selectively reduced PMN-MDSCs in the TME, promoted CD8+ T cell responses, and sensitized tumors to anti-PD-1. TFF2–MSA showed superior antitumor efficacy over existing PMN-targeted strategies. Reduced levels of plasma TFF2 correlated with elevated CXCR4+LOX-1+ neutrophils in patients with GC.

Contributed by Shishir Pant

Qian and Ma et al. fused TFF2, a partial CXCR4 agonist, with murine serum albumin (MSA) to develop TFF2–MSA peptides that restore anti-PD-1 sensitivity in syngeneic gastric cancer (GC) tumor models. TFF2-MSA distinctly modulated CXCR4 signaling, reducing bone marrow granulopoiesis compared to full agonists. TFF2–MSA selectively reduced PMN-MDSCs in the TME, promoted CD8+ T cell responses, and sensitized tumors to anti-PD-1. TFF2–MSA showed superior antitumor efficacy over existing PMN-targeted strategies. Reduced levels of plasma TFF2 correlated with elevated CXCR4+LOX-1+ neutrophils in patients with GC.

Contributed by Shishir Pant

ABSTRACT: Pathologically activated immunosuppressive neutrophils impair cancer immunotherapy efficacy. The chemokine receptor CXCR4, a central regulator of hematopoiesis and neutrophil biology, represents an attractive target. Here, we fuse a secreted CXCR4 partial agonist, trefoil factor 2 (TFF2), to mouse serum albumin (MSA) and demonstrate that TFF2-MSA peptide synergizes with anti-PD-1 to inhibit primary tumor growth and distant metastases and prolongs survival in gastric cancer (GC) mouse models. Using histidine decarboxylase (Hdc)-GFP transgenic mice to track polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) in vivo, we find that TFF2-MSA selectively reduces the Hdc-GFP(+)CXCR4(high) immunosuppressive neutrophils, thereby boosting CD8(+) T cell-mediated tumor killing with anti-PD-1. Importantly, TFF2-MSA reduces bone marrow granulopoiesis, contrasting with CXCR4 antagonism, which fails to confer therapeutic benefits. In GC patients, elevated CXCR4(+)LOX-1(+) low-density neutrophils correlate with lower circulating TFF2 levels. Collectively, our studies introduce a strategy that utilizes CXCR4 partial agonism to restore anti-PD-1 sensitivity by targeting immunosuppressive neutrophils and granulopoiesis.

Author Info: (1) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032

Author Info: (1) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (2) Integrated Diagnostic, Human Health, Health and Biosecurity, CSIRO, Westmead, NSW 2070, Australia. (3) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (4) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA; Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China. (5) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA. (6) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA; Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China. (7) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (8) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (9) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA; Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China. (10) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (11) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (12) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (13) Division of Hematology and Medical Oncology, NYU Langone's Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA. (14) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (15) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (16) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA. (17) Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA. (18) Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA. (19) Department of Microbiology and Immunology, Columbia University, New York, NY 10032, USA. (20) Tonix Pharmaceuticals, Inc., Chatham, NJ 07928, USA. (21) Tonix Pharmaceuticals, Inc., Chatham, NJ 07928, USA. (22) Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA; Columbia University Digestive and Liver Diseases Research Center, Columbia University, New York, NY 10032, USA. Electronic address: tcw21@cumc.columbia.edu.

Prolonged but finite antigen presentation promotes reversible defects of "helpless" memory CD8+ T cells

Van der Heide et al. assessed the memory formation of CD8+ effector T cells in response to pathogens without CD4+ T cell help. In helpless environments, the virus was cleared, but recall responses were limited at early stages. However, recall responses were again present at later stages. Maturation of helpless CD8+ T cells into memory populations took place at a delayed pace due to prolonged viral antigen presentation. As antigen exposure reduced over time, memory formation became similar to that of cells in helped environments.

Van der Heide et al. assessed the memory formation of CD8+ effector T cells in response to pathogens without CD4+ T cell help. In helpless environments, the virus was cleared, but recall responses were limited at early stages. However, recall responses were again present at later stages. Maturation of helpless CD8+ T cells into memory populations took place at a delayed pace due to prolonged viral antigen presentation. As antigen exposure reduced over time, memory formation became similar to that of cells in helped environments.

ABSTRACT: Generation of functional memory CD8(+) T cells typically requires engagement of CD4(+) T cells. In certain acutely resolving infections, however, effector and memory CD8(+) T (Tmem) cell formation appears impervious to the lack of CD4(+) T cell help. Nevertheless, "helpless" CD8(+) Tmem cells may respond poorly upon rechallenge. The origin and long-term fate of helpless CD8(+) Tmem cells remain incompletely understood. Using multiple host-pathogen systems, we demonstrate that helpless effector CD8(+) T cell differentiation was largely normal, with a paradoxical accumulation of TCF1(hi) "memory precursors." However, exposure of CD8(+) T cells to residual antigen impaired the development of the Tmem pool. These defects eventually resolved over time, with full restoration of memory potential and recall capacity. Our findings identify prolonged antigen presentation under helpless conditions as an essential determinant for transient CD8(+) Tmem cell dysfunction in acutely resolving infections and highlight plasticity within the Tmem compartment, with implications for vaccination strategies and beyond.

Author Info: (1) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, US

Author Info: (1) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA. Electronic address: verena.vanderheide@mssm.edu. (2) Department of Microbiology, ISMMS, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, ISMMS, New York, NY 10029, USA; Department of Pharmaceutics, Ghent University, 9000 Ghent, Belgium. (3) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA. (4) Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA. (5) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Human Immune Monitoring Center, ISMMS, New York, NY 10029, USA. (6) Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA. (7) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA. (8) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA. (9) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA. (10) Human Immune Monitoring Center, ISMMS, New York, NY 10029, USA. (11) Human Immune Monitoring Center, ISMMS, New York, NY 10029, USA. (12) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA. (13) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Department of Oncological Sciences, ISMMS, New York, NY 10029, USA; Tisch Cancer Institute, ISMMS, New York, NY 10029, USA. (14) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Department of Microbiology, ISMMS, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, ISMMS, New York, NY 10029, USA; Icahn Genomics Institute, ISMMS, New York, NY 10029, USA. (15) Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA. (16) Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA. (17) Marc and Jennifer Lipschultz Precision Immunology Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA; Diabetes, Obesity and Metabolism Institute, Department of Medicine, ISMMS, New York, NY 10029, USA. Electronic address: dxh2185@miami.edu.

In vivo CAR T cell generation to treat cancer and autoimmune disease

Hunter et al. developed LNPs for in vivo i.v. CAR delivery, with reduced liver tropism and superior tolerability in rats and non-human primates (NHP) compared to Pfizer’s COVID vaccine formulation. In humanized mice and NHPs, CD8-targeted LNPs encapsulating optimized anti-CD19 CAR mRNA facilitated preferential CAR expression in CD8+ T cells (peaked around 6h) and rapidly depleted B cells in blood (within hours) and tissues. After dosing, rebounding NHP B cells were skewed towards a naive phenotype. The LNPs also treated Nalm6 xenograft tumors in PBMC-humanized mice, and generated functional CAR T cells from autoimmune patient T cells.

Contributed by Alex Najibi

Hunter et al. developed LNPs for in vivo i.v. CAR delivery, with reduced liver tropism and superior tolerability in rats and non-human primates (NHP) compared to Pfizer’s COVID vaccine formulation. In humanized mice and NHPs, CD8-targeted LNPs encapsulating optimized anti-CD19 CAR mRNA facilitated preferential CAR expression in CD8+ T cells (peaked around 6h) and rapidly depleted B cells in blood (within hours) and tissues. After dosing, rebounding NHP B cells were skewed towards a naive phenotype. The LNPs also treated Nalm6 xenograft tumors in PBMC-humanized mice, and generated functional CAR T cells from autoimmune patient T cells.

Contributed by Alex Najibi

ABSTRACT: Chimeric antigen receptor (CAR) T cell therapies have transformed treatment of B cell malignancies. However, their broader application is limited by complex manufacturing processes and the necessity for lymphodepleting chemotherapy, restricting patient accessibility. We present an in vivo engineering strategy using targeted lipid nanoparticles (tLNPs) for messenger RNA delivery to specific T cell subsets. These tLNPs reprogrammed CD8(+) T cells in both healthy donor and autoimmune patient samples, and in vivo dosing resulted in tumor control in humanized mice and B cell depletion in cynomolgus monkeys. In cynomolgus monkeys, the reconstituted B cells after depletion were predominantly nave, suggesting an immune system reset. By eliminating the requirements for complex ex vivo manufacturing, this tLNP platform holds the potential to make CAR T cell therapies more accessible and applicable across additional clinical indications.

Author Info: (1) Capstan Therapeutics, San Diego, CA, USA. (2) Capstan Therapeutics, San Diego, CA, USA. (3) Capstan Therapeutics, San Diego, CA, USA. (4) Capstan Therapeutics, San Diego, CA, U

Author Info: (1) Capstan Therapeutics, San Diego, CA, USA. (2) Capstan Therapeutics, San Diego, CA, USA. (3) Capstan Therapeutics, San Diego, CA, USA. (4) Capstan Therapeutics, San Diego, CA, USA. (5) Capstan Therapeutics, San Diego, CA, USA. (6) Capstan Therapeutics, San Diego, CA, USA. (7) Capstan Therapeutics, San Diego, CA, USA. (8) Capstan Therapeutics, San Diego, CA, USA. (9) Capstan Therapeutics, San Diego, CA, USA. (10) Capstan Therapeutics, San Diego, CA, USA. (11) Capstan Therapeutics, San Diego, CA, USA. (12) Capstan Therapeutics, San Diego, CA, USA. (13) Capstan Therapeutics, San Diego, CA, USA. (14) Capstan Therapeutics, San Diego, CA, USA. (15) Capstan Therapeutics, San Diego, CA, USA. (16) Capstan Therapeutics, San Diego, CA, USA. (17) Capstan Therapeutics, San Diego, CA, USA. (18) Capstan Therapeutics, San Diego, CA, USA. (19) Capstan Therapeutics, San Diego, CA, USA. (20) Capstan Therapeutics, San Diego, CA, USA. (21) Capstan Therapeutics, San Diego, CA, USA. (22) Capstan Therapeutics, San Diego, CA, USA. (23) Capstan Therapeutics, San Diego, CA, USA. (24) Capstan Therapeutics, San Diego, CA, USA. (25) Capstan Therapeutics, San Diego, CA, USA. (26) Capstan Therapeutics, San Diego, CA, USA. (27) Capstan Therapeutics, San Diego, CA, USA. (28) Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA. (29) Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA. (30) Capstan Therapeutics, San Diego, CA, USA. (31) Capstan Therapeutics, San Diego, CA, USA. (32) Capstan Therapeutics, San Diego, CA, USA. (33) Capstan Therapeutics, San Diego, CA, USA. (34) Capstan Therapeutics, San Diego, CA, USA. (35) Capstan Therapeutics, San Diego, CA, USA. (36) Capstan Therapeutics, San Diego, CA, USA. (37) Capstan Therapeutics, San Diego, CA, USA. (38) Capstan Therapeutics, San Diego, CA, USA. (39) Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA. Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA. Parker Institute for Cancer Immunotherapy at the University of Pennsylvania, Philadelphia, PA, USA. (40) Capstan Therapeutics, San Diego, CA, USA. Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.

OX40-heparan sulfate binding facilitates CAR T cell penetration into solid tumors in mice

Zhang and Zhong et al. focused on the role of glycosaminoglycans (GAGs) in regulating T cell responses in the TME. In multiple tumor models (pancreatic, breast, lung), a CD20-targeted CAR expressing full length OX40 showed superior antitumor efficacy in an OX40L-dependent and independent manner, suggesting an unidentified OX40L. Whole genome screening revealed that heparan sulfate (HS) and heparin directly bound to and activated OX40, and like OX40L, activated AKT, MAPK, and NFκB signaling. Functionally, the HS–OX40 interaction enhanced cell adhesion and avidity of CAR T cell binding to tumor cells, and increased solid tumor cell penetration.

Contributed by Katherine Turner

Zhang and Zhong et al. focused on the role of glycosaminoglycans (GAGs) in regulating T cell responses in the TME. In multiple tumor models (pancreatic, breast, lung), a CD20-targeted CAR expressing full length OX40 showed superior antitumor efficacy in an OX40L-dependent and independent manner, suggesting an unidentified OX40L. Whole genome screening revealed that heparan sulfate (HS) and heparin directly bound to and activated OX40, and like OX40L, activated AKT, MAPK, and NFκB signaling. Functionally, the HS–OX40 interaction enhanced cell adhesion and avidity of CAR T cell binding to tumor cells, and increased solid tumor cell penetration.

Contributed by Katherine Turner

ABSTRACT: Although chimeric antigen receptor (CAR)-modified T cells have shown great success in treating B cell malignancies, they have demonstrated only limited efficacy against solid tumors. Here, we designed a CAR by integrating an antigen-independent OX40 that showed superior antitumor efficacy against multiple solid tumors. We unexpectedly found, through a CRISPR-Cas9-based whole-genome screen, that heparan sulfate is a ligand for OX40. We found that heparan sulfate can directly bind OX40 at the biochemical and cellular levels and that the interaction of heparan sulfate and OX40 activated the AKT, MAPK, and NF-_B signaling pathways. Functionally, the heparan sulfate-OX40 interaction enhanced cell adhesion and CAR T cell functional binding avidity to tumor cells. In vivo, OX40-expressing CAR T cells exhibited increased solid tumor infiltration and persistence dependent on the OX40-heparan sulfate interaction. Our findings provide insights into a glycan-costimulation interaction that is capable of regulating T cell immunity and has potential application in CAR T cell optimization.

Author Info: (1) Shanghai Lung Cancer Center, Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, C

Author Info: (1) Shanghai Lung Cancer Center, Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China. Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China. (2) Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China. (3) Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China. (4) Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China. (5) Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China. (6) Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China. (7) Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China. (8) Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China. (9) Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China. (10) Shanghai Lung Cancer Center, Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China. (11) Department of Immunology, School of BioMedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia. (12) Department of Immunology, School of BioMedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia. Central Clinical Laboratory, Mongolia Japan Hospital, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia. (13) Department of Immunology, School of BioMedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia. (14) Department of Immunology, School of BioMedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia. (15) Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China. Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China. Joint International Research Laboratory of Metabolic and Developmental Sciences, Shanghai Jiao Tong University, Shanghai 200240, China. Department of Gynaecology and Obstetrics, Shanghai Pudong New Area People's Hospital, Shanghai 200240, China. Engineering Research Center of Cell and Therapeutic Antibody, MOE, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.

Synthetic biology-driven induction of mature TLS formation enhances antitumor immunity in colorectal cancer

Mi et al. developed an attenuated Salmonella typhimurium strain that harbored a quorum-sensing synchronized lysis circuit (SLC) and overexpressed LIGHT. Intragastric SLCVNP20009 treatment colonized tumors to mediate intratumoral lysis and locally release LIGHT. In mouse CRC models, SLCVNP20009 expanded CD4+ and CD8+ cells, cDCs, and group 3 intestinal innate lymphoid cells (ILC3) critical for mucosal host/microbe interactions, promoted formation and maturation of tertiary lymphoid structures (mTLS), and inhibited tumor growth. Fewer mTLSs and reduced survival were observed in tumor-bearing mice deficient in ILC3 or LIGHT receptor HVEM.

Contributed by Paula Hochman

Mi et al. developed an attenuated Salmonella typhimurium strain that harbored a quorum-sensing synchronized lysis circuit (SLC) and overexpressed LIGHT. Intragastric SLCVNP20009 treatment colonized tumors to mediate intratumoral lysis and locally release LIGHT. In mouse CRC models, SLCVNP20009 expanded CD4+ and CD8+ cells, cDCs, and group 3 intestinal innate lymphoid cells (ILC3) critical for mucosal host/microbe interactions, promoted formation and maturation of tertiary lymphoid structures (mTLS), and inhibited tumor growth. Fewer mTLSs and reduced survival were observed in tumor-bearing mice deficient in ILC3 or LIGHT receptor HVEM.

Contributed by Paula Hochman

ABSTRACT: The efficacy of immunotherapy in colorectal cancer (CRC) hinges upon a comprehensive understanding of how the immune system interacts with tumor cells within the colorectal microenvironment. Mature tertiary lymphoid structures (mTLSs) are associated with an increased objective response rate, progression-free survival, and overall survival in patients with CRC. Thus, it has been suggested that increasing mTLSs in the context of CRC could improve patient outcomes. However, no established method to specifically induce TLS maturation within and around tumor sites is available. To address this gap in technology, we engineered a Salmonella typhimurium strain, SLC(VNP20009), to express tumor necrosis factor (TNF) superfamily member 14 (TNFSF14, also called LIGHT). This strain colonized tumors and released LIGHT, which then formed a ligand-receptor pair with herpes virus entry mediator (HVEM) to induce a powerful cellular immune response. Furthermore, this engineered microbe modulated the proportions of intestinal innate lymphoid cells (ILCs), which serve an anti-infection role in innate immunity. Mice that were deficient in HVEM or ILC3 exhibited fewer mTLSs, a greater bacterial burden, and increased mortality in two different models of CRC. Thus, this engineered microbe with enhanced immunogenic properties demonstrated the potential to stimulate mTLS-associated antitumor immune responses in the colon and was well tolerated in vivo. Our results indicate that LIGHT-HVEM signaling on group 3 ILCs (ILC3s) is crucial for mTLS formation and T cell-mediated antitumor immunity in CRC and additionally suggest a synbiotic-based therapeutic approach for the management of CRC.

Author Info: (1) Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China. (2) Department of Radiology, Third Xiangya Hospital, Central South Uni

Author Info: (1) Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China. (2) Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China. (3) Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China. (4) Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China. (5) Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China. (6) Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China. (7) Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China. (8) Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore. Department of Chemical and Biomolecular Engineering and Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore. Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore. Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore. Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore. Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore 138673, Singapore. Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 138667, Singapore. (9) Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China. Key Laboratory of Biological Nanotechnology of National Health Commission, Changsha, Hunan 410008, China. Furong Laboratory, Changsha, Hunan 410008, China.

PSGL-1 is a phagocytosis checkpoint that enables tumor escape from macrophage clearance Featured  

Zhong and Wang et al. found that P-selectin glycoprotein ligand 1 (PSGL-1) was highly expressed in several blood cancers and was associated with poor prognosis. PSGL-1 was found to prevent interactions between ICAM-1 on tumor cells and CD11a/CD18 integrin (LFA-1) on macrophages, limiting macrophage phagocytosis of tumor cells. A humanized antibody targeting human PSGL-1 effectively induced macrophage phagocytosis of human blood cancers in vitro and hindered cancer progression in a variety of mouse models. Anti-PSGL-1 also sensitized tumors to doxorubicin chemotherapy and showed potential synergy with both anti-CD47 and anti-CD38.

Zhong and Wang et al. found that P-selectin glycoprotein ligand 1 (PSGL-1) was highly expressed in several blood cancers and was associated with poor prognosis. PSGL-1 was found to prevent interactions between ICAM-1 on tumor cells and CD11a/CD18 integrin (LFA-1) on macrophages, limiting macrophage phagocytosis of tumor cells. A humanized antibody targeting human PSGL-1 effectively induced macrophage phagocytosis of human blood cancers in vitro and hindered cancer progression in a variety of mouse models. Anti-PSGL-1 also sensitized tumors to doxorubicin chemotherapy and showed potential synergy with both anti-CD47 and anti-CD38.

ABSTRACT: Cancer immunotherapies exhibit impressive efficacy in some cancers but show only limited benefits for refractory hematological malignancies. The complex immune escape mechanisms of hematological cancers remain unclear. Here, we found that P-selectin glycoprotein ligand 1 (PSGL-1) was highly expressed by hematological cancers and negatively correlated with cancer prognosis. PSGL-1 deficiency in tumors suppressed the progression of multiple mouse models of hematological cancer by promoting infiltration of macrophages and their phagocytic activity. Tumor PSGL-1 inhibited the interaction between tumor ICAM-1 and CD11a/CD18 integrin (LFA-1) in macrophages, thereby suppressing prophagocytic signaling downstream of LFA-1. A humanized antibody targeting human PSGL-1 (_hPSGL-1) efficiently triggered macrophage phagocytosis of human hematological malignancies in vitro and slowed cancer progression in vivo. Additionally, PSGL-1 blockade potentiated the efficacy of doxorubicin chemotherapy and anti-CD47 and anti-CD38 antibody therapy. Therefore, PSGL-1 is a previously undescribed phagocytosis checkpoint, and targeting PSGL-1 could be a promising immunotherapy strategy for treating hematological malignancies.

Author Info: (1) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. Jinfeng Laboratory, Chongqing, China. (2) Department of Immun

Author Info: (1) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. Jinfeng Laboratory, Chongqing, China. (2) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. Jinfeng Laboratory, Chongqing, China. (3) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. (4) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. Jinfeng Laboratory, Chongqing, China. (5) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. (6) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. (7) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. (8) GMU-GIBH Joint School of Life Sciences, Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China. (9) College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China. Hangzhou Medimscience Biomedical Technology Co. Ltd., Hangzhou, China. (10) Hangzhou Medimscience Biomedical Technology Co. Ltd., Hangzhou, China. Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China. (11) Molecular Imaging Center, Guangdong Provincial Key Laboratory of Biomedical Imaging, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China. (12) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. Jinfeng Laboratory, Chongqing, China. (13) Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen, China. (14) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. (15) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. (16) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. (17) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. (18) Department of Hematology, Second Xiangya Hospital, Central South University, Changsha, China. (19) Department of Hematology & Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China. (20) Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China. (21) Department of Hematology & Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China. (22) Hangzhou Medimscience Biomedical Technology Co. Ltd., Hangzhou, China. (23) State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China. (24) Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. Jinfeng Laboratory, Chongqing, China. Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China. Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.

Armored human CAR T(reg) cells with PD1 promoter-driven IL-10 have enhanced suppressive function Spotlight 

Boardman and Mangat et al. asked if engineering CAR Treg cells to enhance a suppressive mechanism (IL-10) and simultaneously remove an inhibitory signal (PD-1) would improve their suppressive function. CRISPR-mediated PD-1 deletion increased CAR Treg cell activation, while knock-in of IL-10 into the PDCD1 locus of HLA-A2-specific CAR Tregs (IL-10KITreg), resulted in high levels of antigen-dependent, CAR-regulated IL-10 secretion. IL-10KITreg cells demonstrated increased suppression of dendritic cells and allo-antigen- and islet auto-antigen-specific T cells. In vivo, IL-10KITreg cells were safe, stable, secreted IL-10, and suppressed DCs and xenogeneic GvHD.

Contributed by Katherine Turner

Boardman and Mangat et al. asked if engineering CAR Treg cells to enhance a suppressive mechanism (IL-10) and simultaneously remove an inhibitory signal (PD-1) would improve their suppressive function. CRISPR-mediated PD-1 deletion increased CAR Treg cell activation, while knock-in of IL-10 into the PDCD1 locus of HLA-A2-specific CAR Tregs (IL-10KITreg), resulted in high levels of antigen-dependent, CAR-regulated IL-10 secretion. IL-10KITreg cells demonstrated increased suppression of dendritic cells and allo-antigen- and islet auto-antigen-specific T cells. In vivo, IL-10KITreg cells were safe, stable, secreted IL-10, and suppressed DCs and xenogeneic GvHD.

Contributed by Katherine Turner

ABSTRACT: Regulatory T cell (T(reg) cell) therapy has been transformed through the use of chimeric antigen receptors (CARs). We previously found that human T(reg) cells minimally produce IL-10 and have a limited capacity to control innate immunity compared to type 1 regulatory T cells (T(r)1 cells). To create "hybrid" CAR T(reg) cells with T(r)1 cell-like properties, we examined whether the PDCD1 locus could be exploited to endow T(reg) cells with CAR-regulated IL-10 expression. CRISPR-mediated PD1 deletion increased CAR T(reg) cell activation, and knock-in of IL10 under control of the PD1 promoter resulted in CAR-induced IL-10 secretion. IL10 knock-in improved CAR T(reg) cell function, as determined by increased suppression of dendritic cells and alloantigen- and islet autoantigen-specific T cells. In vivo, IL10 knock-in CAR T(reg) cells were stable, safe, and suppressed dendritic cells and xenogeneic graft-versus-host disease. CRISPR-mediated engineering to simultaneously remove an inhibitory signal and enhance a suppressive mechanism is a previously unexplored approach to improve CAR T(reg) cell potency.

Author Info: (1) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (2) Department of Surgery, T

Author Info: (1) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (2) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada. (3) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (4) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (5) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (6) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (7) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (8) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (9) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (10) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (11) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (12) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. (13) Department of Pathology, Stanford University, Stanford, CA, USA. (14) Department of Surgery, The University of British Columbia, Vancouver, BC, Canada. BC Children's Hospital Research Institute, Vancouver, BC, Canada. School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada.

First-in-Human Clinical Trial of Vaccination with WDVAX, a Dendritic Cell Activating Scaffold Incorporating Autologous Tumor Cell Lysate, in Metastatic Melanoma Patients Spotlight 

Based on a first-in-human phase I trial in patients with metastatic melanoma, Hodi et al. reported safety and feasibility of WDVAX, a macroporous poly-lactide-coglycolide matrix polymer scaffold incorporating cytokine GM-CSF, the innate Toll-like receptor 9 agonist CpG oligonucleotide, and autologous tumor lysate. WDVAX showed a favorable safety profile and manufacturing feasibility. Nine patients (42.9%) had stable disease as BOR, with an encouraging 12-month survival estimate of 94%, and a median time-to-progression of 12.4 months. The immune assessments support immunogenicity of the vaccine, with heterogeneous induction of T cells and myeloid cells.

Contributed by Shishir Pant

Based on a first-in-human phase I trial in patients with metastatic melanoma, Hodi et al. reported safety and feasibility of WDVAX, a macroporous poly-lactide-coglycolide matrix polymer scaffold incorporating cytokine GM-CSF, the innate Toll-like receptor 9 agonist CpG oligonucleotide, and autologous tumor lysate. WDVAX showed a favorable safety profile and manufacturing feasibility. Nine patients (42.9%) had stable disease as BOR, with an encouraging 12-month survival estimate of 94%, and a median time-to-progression of 12.4 months. The immune assessments support immunogenicity of the vaccine, with heterogeneous induction of T cells and myeloid cells.

Contributed by Shishir Pant

ABSTRACT: The optimal means to prime for effective anti-tumor immunity in a cancer patient remains elusive in the current era of checkpoint blockade. Crafting a strategy to amplify CD8+ T cells while blocking regulatory cells should increase immunotherapy efficacy. Biomaterial carriers have been demonstrated in preclinical studies to amplify the effects of immunomodulatory agents, synergistically integrate the effects of different agents, and concentrate and manipulate immune cells in vivo. In this phase I trial in patients with metastatic melanoma, the cytokine GM-CSF and the innate TLR9 agonist CpG oligonucleotide were admixed with autologous tumor lysate onto a microporous poly-lactide-co-glycolide (PLG) matrix polymer scaffold that achieves precise control over the spatial and temporal release of immunostimulatory agents in vivo. This materials system served as a physical antigen-presenting structure for which dendritic cells and other immune-stimulating cells are recruited and activated (WDVAX). In this first clinical trial of a macroscale biomaterial-based vaccine, WDVAX treatment was found to be feasible and induced immune activation in melanoma patients.

Author Info: (1) Dana-Farber Cancer Institute, Boston, Massachusetts, United States. (2) Dana-Farber Cancer Institute, Boston, MA, United States. (3) Wyss Institute for Biologically Inspired En

Author Info: (1) Dana-Farber Cancer Institute, Boston, Massachusetts, United States. (2) Dana-Farber Cancer Institute, Boston, MA, United States. (3) Wyss Institute for Biologically Inspired Engineering, United States. (4) Dana-Farber Cancer Institute, Boston, United States. (5) Dana-Farber Cancer Institute, United States. (6) Dana-Farber Cancer Institute, Boston, MA, United States. (7) Dana-Farber Cancer Institute, United States. (8) Dana-Farber Cancer Institute, Boston, MA, United States. (9) Dana-Farber Cancer Institute, Boston, MA, United States. (10) Dana-Farber Cancer Institute, Boston, MA, United States. (11) Attivare Therapeutics Inc., Natick, MA, United States. (12) Wyss Institute for Biologically Inspired Engineering, Boston, United States. (13) Dana-Farber Cancer Institute, Boston, MA, United States. (14) Dana-Farber Cancer Institute, Boston, MA, United States. (15) Dana-Farber Cancer Institute, Boston, United States. (16) Dana-Farber Cancer Institute, Boston, MA, United States. (17) Dana-Farber Cancer Institute, Boston, MA, United States. (18) Brigham and Women's Hospital, Boston, MA, United States. (19) Dana-Farber Cancer Institute, Boston, Massachusetts, United States. (20) Harvard University, Cambridge, MA, United States.

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.