Journal Articles

Radiotherapy enhances anticancer CD8 T cell responses by cGAMP transfer through LRRC8A/C volume-regulated anion channels Spotlight 

Cao et al. demonstrated that the LRRC8A/C volume-regulated anion channel (VRAC) transfers cGAMP from irradiated cancer cells into immune cells within the tumor, and enhances their effector function. TCR signaling opened VRAC pores to facilitate cGAMP import, leading to STING activation, interferon-α/β induction, and upregulation of granzymes and IFNγ in CD8+ T cells. ENPP1 and CD39 blockade maintained extracellular ATP and cGAMP levels. and further supported VRAC-mediated CD8+ T cell activation. Human TCGA analysis revealed that high LRRC8A/C correlated with enhanced STING signaling, T cell responses, and improved OS in various cancers.

Contributed by Shishir Pant

Cao et al. demonstrated that the LRRC8A/C volume-regulated anion channel (VRAC) transfers cGAMP from irradiated cancer cells into immune cells within the tumor, and enhances their effector function. TCR signaling opened VRAC pores to facilitate cGAMP import, leading to STING activation, interferon-α/β induction, and upregulation of granzymes and IFNγ in CD8+ T cells. ENPP1 and CD39 blockade maintained extracellular ATP and cGAMP levels. and further supported VRAC-mediated CD8+ T cell activation. Human TCGA analysis revealed that high LRRC8A/C correlated with enhanced STING signaling, T cell responses, and improved OS in various cancers.

Contributed by Shishir Pant

ABSTRACT: The volume-regulated anion channels (VRACs) transport osmolytes, neurotransmitters, and cyclic GMP-AMP (cGAMP) across the cell membrane to regulate cell volume and host defense. We report that the leucine-rich repeat-containing 8A/C (LRRC8A/C) VRAC plays a crucial role in immune responses to radiotherapy and chemotherapy for cancer. VRACs transfer cGAMP from irradiated cancer cells to infiltrating CD4 and CD8 T cells, thus enhancing their effector functions. TCR signaling acts as a physiological signal to open the VRAC pore through phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] and reactive oxygen species (ROS). This allows the rapid uptake of cGAMP and STING activation in mouse and human T cells and induction of interferon-α/β, which up-regulate granzymes and IFN-γ in CD8 T cells. Inhibition of the extracellular hydroxylases CD39 and ENPP1 maintains extracellular ATP and cGAMP, which promotes VRAC-enhanced CD8 T cell anticancer function. Thus, the transfer of cGAMP to T cells by VRACs may be a strategy that can be targeted in future cancer therapies.

Author Info: (1) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences,

Author Info: (1) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (2) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200092, China. (3) State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China. (4) Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China. (5) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (6) Center for Allergic and Inflammatory Diseases and Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai 200031, China. CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (7) State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China. (8) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (9) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, China. (10) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (11) Center for Allergic and Inflammatory Diseases and Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai 200031, China. (12) Center for Allergic and Inflammatory Diseases and Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai 200031, China. (13) School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China. (14) Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China. (15) CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (16) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (17) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (18) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (19) Suzhou Blood Center, Suzhou 215000, Jiangsu, China. (20) Suzhou Blood Center, Suzhou 215000, Jiangsu, China. (21) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (22) CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (23) CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (24) State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China. (25) Shenzhen Medical Academy of Research and Translation, Shenzhen 518107, Guangdong, China. (26) Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, China. (27) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China. (28) State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 200437, China. (29) Departments of Physiology, Neuroscience, and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. (30) School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China. (31) School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China. (32) Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China. (33) Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200092, China. (34) Center for Allergic and Inflammatory Diseases and Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai 200031, China. (35) State Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.

Proximity between LAG-3 and the T cell receptor guides suppression of T cell activation and autoimmunity Spotlight 

Du, Chen, and You et al. explored mechanisms of LAG3-mediated T cell suppression. Optimal LAG3 inhibitory function required spatial proximity of LAG3 to TCRs (but not the CD4 co-receptor), and was facilitated by cognate peptide–MHC-II binding, resulting in disruption of intracellular CD3ε/Lck association via LAG3’s FSAL motif. To maximize the inhibitory effect of LAG3, a bridging LAG3/TCR bispecific mAb was developed to enforce LAG3/TCR proximity, which potently suppressed both CD4+ and CD8+ T cell responses and was therapeutically effective in mouse autoimmune models of multiple sclerosis, type 1 diabetes, and hepatitis.

Contributed by Katherine Turner

Du, Chen, and You et al. explored mechanisms of LAG3-mediated T cell suppression. Optimal LAG3 inhibitory function required spatial proximity of LAG3 to TCRs (but not the CD4 co-receptor), and was facilitated by cognate peptide–MHC-II binding, resulting in disruption of intracellular CD3ε/Lck association via LAG3’s FSAL motif. To maximize the inhibitory effect of LAG3, a bridging LAG3/TCR bispecific mAb was developed to enforce LAG3/TCR proximity, which potently suppressed both CD4+ and CD8+ T cell responses and was therapeutically effective in mouse autoimmune models of multiple sclerosis, type 1 diabetes, and hepatitis.

Contributed by Katherine Turner

ABSTRACT: Therapeutically targeting pathogenic T cells in autoimmune diseases has been challenging. Although LAG-3, an inhibitory checkpoint receptor specifically expressed on activated T cells, is known to bind to major histocompatibility complex class II (MHC class II), we demonstrate that MHC class II interaction alone is insufficient for optimal LAG-3 function. Instead, LAG-3's spatial proximity to T cell receptor (TCR) but not CD4 co-receptor, facilitated by cognate peptide-MHC class II, is crucial in mediating CD4(+) T cell suppression. Mechanistically, LAG-3 forms condensate with TCR signaling component CD3_ through its intracellular FSAL motif, disrupting CD3_/lymphocyte-specific protein kinase (Lck) association. To exploit LAG-3's proximity to TCR and maximize LAG-3-dependent T cell suppression, we develop an Fc-attenuated LAG-3/TCR inhibitory bispecific antibody to bypass the requirement of cognate peptide-MHC class II. This approach allows for potent suppression of both CD4(+) and CD8(+) T cells and effectively alleviates autoimmune symptoms in mouse models. Our findings reveal an intricate and conditional checkpoint modulatory mechanism and highlight targeting of LAG-3/TCR cis-proximity for T cell-driven autoimmune diseases lacking effective and well-tolerated immunotherapies.

Author Info: (1) Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Hea

Author Info: (1) Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA. (2) State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China. (3) Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA. (4) Department of Cardiology and Department of Cell Biology, The Second Affiliated Hospital, Zhejiang University School of Medicine, and Liangzhu Laboratory, Zhejiang University, Hangzhou 310012, Zhejiang, China; Kidney Disease Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China. (5) Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA. (6) Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA. (7) University of Chinese Academy of Sciences, Beijing 100049, China. (8) Department of Cardiology and Department of Cell Biology, The Second Affiliated Hospital, Zhejiang University School of Medicine, and Liangzhu Laboratory, Zhejiang University, Hangzhou 310012, Zhejiang, China. (9) Hansjšrg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA. (10) Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA. (11) Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA. (12) Microscopy Core, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY 10016, USA. (13) Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, Zhejiang, China. (14) The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA. (15) Hansjšrg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA. (16) Department of Cardiology and Department of Cell Biology, The Second Affiliated Hospital, Zhejiang University School of Medicine, and Liangzhu Laboratory, Zhejiang University, Hangzhou 310012, Zhejiang, China. Electronic address: jackweichen@zju.edu.cn. (17) State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China. Electronic address: jlou@ibp.ac.cn. (18) Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA. Electronic address: jun.wang@nyulangone.org.

Engineered CD4 TCR T cells with conserved high-affinity TCRs targeting NY-ESO-1 for advanced cellular therapies in cancer Spotlight 

Saillard et al. demonstrated the clinical potential of TCR-engineered CD4+ T cells targeting a broad range of NY-ESO-1–expressing tumors in HLA-DRB3*02:02-positive cancers. HLA-DRB3*02:02–restricted NYESO-1-specific CD4+ T cells exhibited high cytotoxicity, strong T helper 1 polarization, and a dominant TCRαβ usage across patients and anatomical sites. These cytotoxic TCR-biased CD4+ T cells were also present in other NY-ESO-1-expressing cancers. Adoptive T cell therapy using engineered CD4+ T cells with these conserved TCR chain pairs showed robust antigen-specific cytotoxicity in vitro and in vivo against HLA-matched xenograft tumors.

Contributed by Shishir Pant

Saillard et al. demonstrated the clinical potential of TCR-engineered CD4+ T cells targeting a broad range of NY-ESO-1–expressing tumors in HLA-DRB3*02:02-positive cancers. HLA-DRB3*02:02–restricted NYESO-1-specific CD4+ T cells exhibited high cytotoxicity, strong T helper 1 polarization, and a dominant TCRαβ usage across patients and anatomical sites. These cytotoxic TCR-biased CD4+ T cells were also present in other NY-ESO-1-expressing cancers. Adoptive T cell therapy using engineered CD4+ T cells with these conserved TCR chain pairs showed robust antigen-specific cytotoxicity in vitro and in vivo against HLA-matched xenograft tumors.

Contributed by Shishir Pant

ABSTRACT: While cancer immunotherapy has primarily focused on CD8 T cells, CD4 T cells are increasingly recognized for their role in antitumor immunity. The HLA-DRB3*02:02 allele is found in 50% of Caucasians. In this study, we screened HLA-DRB3*02:02 patients with melanoma for tumor-specific CD4 T cells and identified robust New York esophageal squamous cell carcinoma 1 (NY-ESO-1)123-137/HLA-DRB3*02:02 CD4 T cell activity in both peripheral blood and tumor tissue. By analyzing NY-ESO-1123-137/HLA-DRB3*02:02-restricted CD4 T cell clones, we uncovered an unexpectedly high cytotoxicity, strong T helper 1 polarization, and recurrent αβ T cell receptor (TCRαβ) usage across patients and anatomical sites. These responses were also present in other NY-ESO-1-expressing cancers. TCRs from these clones, when transduced into primary CD4 T cells, showed direct antitumor efficacy both in vitro and in vivo. Our findings suggest that these TCRs are promising for adoptive T cell transfer therapy, enabling broader targeting of NY-ESO-1-expressing adult and pediatric cancers in clinical settings.

Author Info: (1) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. Department of Pathology and Immuno

Author Info: (1) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland. Translational Research Centre in Onco-Hematology (CRTOH), University of Geneva, Geneva, Switzerland. Geneva Centre for Inflammation Research (GCIR), University of Geneva, Geneva, Switzerland. (2) Ludwig Institute for Cancer Research, Lausanne, Switzerland. Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland. Translational Research Centre in Onco-Hematology (CRTOH), University of Geneva, Geneva, Switzerland. Geneva Centre for Inflammation Research (GCIR), University of Geneva, Geneva, Switzerland. (3) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (4) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (5) Ludwig Institute for Cancer Research, Lausanne, Switzerland. Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland. Translational Research Centre in Onco-Hematology (CRTOH), University of Geneva, Geneva, Switzerland. Geneva Centre for Inflammation Research (GCIR), University of Geneva, Geneva, Switzerland. (6) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (7) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (8) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (9) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (10) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (11) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland. (12) Division of Nephrology, Geneva University Hospitals, Geneva, Switzerland. Division of Transplantation Immunology, Geneva University Hospitals, Geneva, Switzerland. (13) Pediatric Hematology-Oncology Unit, Division of Pediatrics, Department "Woman-Mother-Child," University Hospital and Lausanne University, Lausanne, Switzerland. (14) Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland. Department of Oncology, Geneva University Hospitals, Geneva, Switzerland. (15) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland. (16) Plateforme clinique et translationnelle, Centre des thŽrapies expŽrimentales, University Hospital and Lausanne University, Lausanne, Switzerland. (17) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (18) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. (19) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (20) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland. (21) UniversitŽ Bourgogne Franche-ComtŽ, INSERM, EFS BFC, UMR1098, Interactions H™te-Greffon-Tumeur/IngŽnierie Cellulaire et GŽnique, Besanon, France. (22) Pediatric Hematology-Oncology Unit, Division of Pediatrics, Department "Woman-Mother-Child," University Hospital and Lausanne University, Lausanne, Switzerland. (23) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (24) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (25) Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland. Translational Research Centre in Onco-Hematology (CRTOH), University of Geneva, Geneva, Switzerland. Geneva Centre for Inflammation Research (GCIR), University of Geneva, Geneva, Switzerland.

GPC2-targeted CAR T cells engineered with NFAT-inducible membrane-tethered IL-15/IL-21 exhibit enhanced activity against neuroblastoma Spotlight 

Okada et al. engineered and evaluated the efficacy of GPC2-CAR T cells with constitutive production of soluble IL-15/IL-21, membrane-tethered IL-15 and IL-21, or NFAT-inducible expression of membrane-tethered IL-15 and IL-21 in a high-tumor-burden neuroblastoma (NB) murine model. All engineered GPC2-CAR T cells showed enhanced cytotoxicity relative to control GPC2-CAR T cells. Compared to CAR T cells with constitutive cytokine expression, which maintained a pro-inflammatory signature after tumor elimination, leading to hypercytokinemia and death, NFAT-inducible tethered IL-15/IL-21 showed an immunosuppressive transcriptional profile with acceptable tolerability.

Contributed by Shishir Pant

Okada et al. engineered and evaluated the efficacy of GPC2-CAR T cells with constitutive production of soluble IL-15/IL-21, membrane-tethered IL-15 and IL-21, or NFAT-inducible expression of membrane-tethered IL-15 and IL-21 in a high-tumor-burden neuroblastoma (NB) murine model. All engineered GPC2-CAR T cells showed enhanced cytotoxicity relative to control GPC2-CAR T cells. Compared to CAR T cells with constitutive cytokine expression, which maintained a pro-inflammatory signature after tumor elimination, leading to hypercytokinemia and death, NFAT-inducible tethered IL-15/IL-21 showed an immunosuppressive transcriptional profile with acceptable tolerability.

Contributed by Shishir Pant

ABSTRACT: Neuroblastoma (NB) is a highly aggressive childhood solid tumor with poor outcomes. Chimeric antigen receptor (CAR) T cells have shown limited efficacy in NB, with the best outcomes reported in patients with a low tumor burden, highlighting the need for further CAR optimization. One approach to addressing the high tumor burden involves engineering CAR T cells to release or express transgenic cytokines. However, its systemic toxicity remains an important therapeutic challenge. Here, we evaluated the efficacy of interleukin (IL)-15- and IL-21-enhanced glypican-2 (GPC2)-targeted CAR T cells (GPC2-CAR T cells) in targeting high-burden NB. Three strategies for expressing the cytokines were evaluated: constitutive secretion (GPC2-CAR+sol.IL15.IL21), constitutive membrane-tethered expression (GPC2-CAR+teth.IL15.IL21), and NFAT-inducible membrane-tethered expression (GPC2-CAR+NFAT.IL15.IL21). Engineered GPC2-CAR T cells were tested in vitro and in vivo using high NB-burden xenograft models. Additionally, single-cell RNA sequencing was used to profile the effector cells in the tumor microenvironment. All three versions of GPC2-CAR T cells significantly enhanced killing against a high NB burden, both in vitro and in vivo, relative to control GPC2-CAR T cells. Mice treated with GPC2-CAR+NFAT.IL15.IL21 exhibited significantly lower anorexia-associated morbidity/mortality. Supporting these data, tumor-infiltrating GPC2-CAR+NFAT.IL15.IL21 developed an immunosuppressive transcriptional profile upon tumor regression, leading to prolonged survival in treated mice. In contrast, GPC2-CAR+teth.IL15.IL21 maintained a pro-inflammatory transcriptional signature despite near tumor clearance, resulting in hypercytokinemia and death. NFAT-inducible co-expression of tethered IL-15/IL-21 enhanced GPC2-CAR T-cell function against a high NB burden with acceptable tolerability in mice. Further studies are required to validate these findings.

Author Info: (1) National Cancer Institute, Bethesda, United States. (2) National Cancer Institute, Bethesda, United States. (3) National Cancer Institute, Bethesda, Maryland, United States. (4

Author Info: (1) National Cancer Institute, Bethesda, United States. (2) National Cancer Institute, Bethesda, United States. (3) National Cancer Institute, Bethesda, Maryland, United States. (4) Keio University School of Medicine, Shinjuku, Tokyo, Japan. (5) National Cancer Institute, Bethesda, United States. (6) National Cancer Institute, Bethesda, MD, United States. (7) NCI-Frederick, Bethesda, MD, United States. (8) National Institutes of Health, United States. (9) National Cancer Institute, Bethesda, MD, United States. (10) National Cancer Institute, Bethesda, MD, United States. (11) National Cancer Institute, Bethesda, Maryland, United States. (12) Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States. (13) National Cancer Institute, Bethesda, MD, United States. (14) National Cancer Institute, Bethesda, MD, United States. (15) National Cancer Institute, Bethesda, United States.

Mutant p53 exploits enhancers to elevate immunosuppressive chemokine expression and impair immune checkpoint inhibitors in pancreatic cancer Featured  

Mahat et al. studied whether p53 mutant proteins that lose their wild-type tumor-suppressor functions might also have potential gains in function, and found that commonly mutated p53R172H, while unable to bind to canonical p53 targets, maintained its transactivating capacity and, facilitated by NF-κB, bound to distal enhancers for Cxcl1. This drove increased expression of Cxcl1, which promoted neutrophil recruitment and an immunosuppressive TME that supported tumor progression. Deletion of mutant p53 enhanced the immune TME, delayed tumor growth, and increased the efficacy of immune checkpoint inhibitors.

Mahat et al. studied whether p53 mutant proteins that lose their wild-type tumor-suppressor functions might also have potential gains in function, and found that commonly mutated p53R172H, while unable to bind to canonical p53 targets, maintained its transactivating capacity and, facilitated by NF-κB, bound to distal enhancers for Cxcl1. This drove increased expression of Cxcl1, which promoted neutrophil recruitment and an immunosuppressive TME that supported tumor progression. Deletion of mutant p53 enhanced the immune TME, delayed tumor growth, and increased the efficacy of immune checkpoint inhibitors.

ABSTRACT: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer characterized by activating KRAS mutations and TP53 alterations. TP53 missense mutations lose their wild-type tumor-suppressor function. Here, we studied whether p53 missense mutations have potential gain-of-function oncogenic roles and their impact on cancer-cell-intrinsic gene expression and the tumor immune microenvironment (TME) in PDAC. p53R172H established an immunosuppressive TME and impaired the efficacy of immune checkpoint inhibitors (ICIs) by regulating a distinct set of chemokines. Among these, tumor-specific reduction of Cxcl1, which encodes a chemoattractant for neutrophils, promoted T cell infiltration and decreased tumor growth. Mechanistically, p53R172H occupied the distal enhancers of Cxcl1 and amplified its expression. These enhancers were responsible for Cxcl1 expression and were essential for its immunosuppressive function. Nuclear factor κB (NF-κB) was a critical cofactor required for p53R172H occupancy at these enhancers. Thus, a common mutation in a tumor-suppressor transcription factor appropriates enhancers, thereby stimulating chemokine expression and establishing an immunosuppressive TME that diminishes ICI efficacy in PDAC.

Author Info: (1) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Electronic address: mahat@mit.edu. (2) Edwin L. Steele Laborato

Author Info: (1) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Electronic address: mahat@mit.edu. (2) Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. (3) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (4) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (5) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (6) Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. (7) Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. (8) Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. (9) Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA. (10) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (11) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (12) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (13) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (14) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (15) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (16) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (17) Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA. (18) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA. (19) Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. Electronic address: rjain@mgh.harvard.edu. (20) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Electronic address: sharppa@mit.edu.

Intratumoral IL12 mRNA administration activates innate and adaptive pathways in checkpoint inhibitor-resistant tumors resulting in complete responses Spotlight 

Lakshmipathi et al. showed that β2-microglobulin (B2M)-KO (MHC-I non-expressing) melanoma and CRC mouse tumor lines grew rapidly and were anti-PD-L1-resistant when engrafted into immune-competent syngeneic mice. I.t. monotherapy with LNP-formulated murine IL-12 mRNA caused CRs in ≥ 60% of both ICI-sensitive (WT) and -resistant (B2M KO) tumors, and enhanced efficacy in the ICI-responsive model. Responses were durable, and cured mice were resistant to rechallenge. Monotherapy induced proinflammatory/TH1 cytokines and chemokines in serum and in the TME, reduced Treg numbers, and increased the numbers and activation state of TAMs and CTLs.

Contributed by Paula Hochman

Lakshmipathi et al. showed that β2-microglobulin (B2M)-KO (MHC-I non-expressing) melanoma and CRC mouse tumor lines grew rapidly and were anti-PD-L1-resistant when engrafted into immune-competent syngeneic mice. I.t. monotherapy with LNP-formulated murine IL-12 mRNA caused CRs in ≥ 60% of both ICI-sensitive (WT) and -resistant (B2M KO) tumors, and enhanced efficacy in the ICI-responsive model. Responses were durable, and cured mice were resistant to rechallenge. Monotherapy induced proinflammatory/TH1 cytokines and chemokines in serum and in the TME, reduced Treg numbers, and increased the numbers and activation state of TAMs and CTLs.

Contributed by Paula Hochman

ABSTRACT: Despite the proven clinical activity of checkpoint inhibitors (ICIs) in several cancer indications, frequent occurrence of primary and secondary resistance reduces their overall effectiveness. Development of ICI resistance has been attributed mainly to genetic or epigenic alterations that affect the tumor antigen presentation machinery leading to diminished anti-tumor immune responses. There is an urgent need for new approaches which can either re-sensitize resistant tumors to the ICIs or engage alternate immune pathways to inhibit tumors. Intratumoral delivery of nanoparticle encapsulated murine IL12 (mIL12) mRNA induces powerful anti-tumor immune responses in murine tumor models, and the human version of this drug results in objective responses in patients with advanced disease. Here, we tested the efficacy of mIL12 mRNA as a single agent and in combination with anti-PD-L1 antibodies in ICI-sensitive Yummer 1.7 melanoma and MC38 colorectal murine tumors and in ICI resistant, _2-microglobulin (B2M) knockout versions of these models. mIL12 mRNA monotherapy was sufficient to cause complete responses (CRs) in_³_60% of both ICI-sensitive or -resistant Yummer 1.7 melanoma and MC38 colorectal carcinoma tumors. The mIL12 mRNA treatment resulted in potent upregulation of T(H)1 type cytokines and chemokines. A reduction in number of Tregs, increase in numbers and activation state of both cytotoxic T cells (CTLs) as well as tumor-associated macrophages (TAMs) was observed indicating enhanced anti-tumor, cell-based immune responses in the tumor microenvironment. This mIL12-induced concerted immune activation was associated with a robust killing and phagocytosis of tumor cells resulting in durable CRs. These observations suggest that intratumoral IL12mRNA therapy may benefit patients with ICI-resistant cancers.

Author Info: (1) Yale Center for Precision Cancer Modeling, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA. (2) Yale Center for Precision Cancer Modeling, Yale Cancer Center, Y

Author Info: (1) Yale Center for Precision Cancer Modeling, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA. (2) Yale Center for Precision Cancer Modeling, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA. (3) Yale Center for Precision Cancer Modeling, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA. (4) Yale Center for Precision Cancer Modeling, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA. (5) Department of Dermatology, Yale School of Medicine, New Haven, CT, USA. Department of Pathology, McGill University, Montreal, Canada. (6) AstraZeneca, Cambridge Biomedical Campus, Cambridge, CB2 0AA, UK. (7) Departments of Pathology and Internal Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA. (8) Yale Center for Precision Cancer Modeling, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA. Departments of Pathology and Internal Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA. Department of Dermatology, Yale School of Medicine, New Haven, CT, USA. Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA. (9) AstraZeneca, Cambridge Biomedical Campus, Cambridge, CB2 0AA, UK. (10) Yale Center for Precision Cancer Modeling, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA. vish.muthusamy@yale.edu. Department of Medicine (Oncology), Yale School of Medicine, New Haven, CT, USA. vish.muthusamy@yale.edu.

Subclonal immune evasion in non-small cell lung cancer Spotlight 

Dijkstra et al. generated patient-derived organoid sublines from spatially distinct tumor regions and separate clones within individual regions of 3 NSCLC samples, and also expanded tumor-reactive autologous TILs or donor-derived NK cells. Tumor cell subpopulations from the same patient showed substantial heterogeneity in their capacity to elicit a T cell or NK cell response, both within and across tumor regions. Genetically distinct subclones represented immune-evasive and non-evasive subpopulations through both antigen-dependent and -independent mechanisms, but these were not predictable based on genetic or transcriptomic features.

Contributed by Shishir Pant

Dijkstra et al. generated patient-derived organoid sublines from spatially distinct tumor regions and separate clones within individual regions of 3 NSCLC samples, and also expanded tumor-reactive autologous TILs or donor-derived NK cells. Tumor cell subpopulations from the same patient showed substantial heterogeneity in their capacity to elicit a T cell or NK cell response, both within and across tumor regions. Genetically distinct subclones represented immune-evasive and non-evasive subpopulations through both antigen-dependent and -independent mechanisms, but these were not predictable based on genetic or transcriptomic features.

Contributed by Shishir Pant

ABSTRACT: Cancers rarely respond completely to immunotherapy. While tumors consist of multiple genetically distinct clones, whether this affects the potential for immune escape remains unclear due to an inability to isolate and propagate individual subclones from human cancers. Here, we leverage the multi-region TRACERx lung cancer evolution study to generate a patient-derived organoid - T cell co-culture platform that allows the functional analysis of subclonal immune escape at single clone resolution. We establish organoid lines from 11 separate tumor regions from three patients, followed by isolation of 81 individual clonal sublines. Co-culture with tumor infiltrating lymphocytes (TIL) or natural killer (NK) cells reveals cancer-intrinsic and subclonal immune escape in all 3 patients. Immune evading subclones represent genetically distinct lineages with a unique evolutionary history. This indicates that immune evading and non-evading subclones can be isolated from the same tumor, suggesting that subclonal tumor evolution directly affects immune escape.

Author Info: (1) Department of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Cancer Evolution and Genome Instability Laboratory, The Francis C

Author Info: (1) Department of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Oncode Institute, Utrecht, the Netherlands. Electronic address: k.dijkstra@nki.nl. (2) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Tumor ImmunoGenomics and Immunosurveillance Laboratory, University College London Cancer Institute, University College London, London, UK. (3) Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Cancer Immunology Unit, Immune Regulation and Tumor Immunotherapy Group, Research Department of Haematology, University College London Cancer Institute, London, UK. (4) Department of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (5) Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Cancer Immunology Unit, Immune Regulation and Tumor Immunotherapy Group, Research Department of Haematology, University College London Cancer Institute, London, UK; Immune Regulation Laboratory, Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom. (6) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (7) Cancer Immunology Unit, Immune Regulation and Tumor Immunotherapy Group, Research Department of Haematology, University College London Cancer Institute, London, UK. (8) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (9) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK. (10) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, Great Ormond Street UCL Institute of Child Health, University College London, London, UK. (11) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (12) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (13) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (14) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (15) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (16) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK. (17) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Tumor ImmunoGenomics and Immunosurveillance Laboratory, University College London Cancer Institute, University College London, London, UK; Division of Medicine, University College London, London, UK. (18) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK. (19) Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Department of Cellular Pathology, University College London Hospital, London, UK. (20) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (21) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (22) Department of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (23) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (24) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (25) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (26) Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Cancer Immunology Unit, Immune Regulation and Tumor Immunotherapy Group, Research Department of Haematology, University College London Cancer Institute, London, UK. (27) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK. (28) Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK; Department of Medical Oncology, University College London Hospitals, London, UK. (29) Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK. (30) Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Tumor ImmunoGenomics and Immunosurveillance Laboratory, University College London Cancer Institute, University College London, London, UK. (31) Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Pre-Cancer Immunology Laboratory, University College London Cancer Institute, London, UK. (32) University College London Cancer Institute, London, UK; Division of Infection and Immunity, University College London, London, UK. (33) Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Cancer Immunology Unit, Immune Regulation and Tumor Immunotherapy Group, Research Department of Haematology, University College London Cancer Institute, London, UK. Electronic address: s.quezada@ucl.ac.uk. (34) Department of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands. Electronic address: e.voest@nki.nl. (35) Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; Department of Medical Oncology, University College London Hospitals, London, UK. Electronic address: charles.swanton@crick.ac.uk.

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

Spotlight 

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 Spotlight 

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 Spotlight 

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.

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.