Journal Articles

Pseudomonas aeruginosa induces tumor pyroptosis and immune activation to enhance checkpoint blockade in colorectal cancer Spotlight 

Hu et al. demonstrated that Pseudomonas aeruginosa (P.a) initiates caspase-3-dependent apoptosis in MC38 CRC cell lines. Pyroptosis – characterized by GSDME cleavage, intracellular ROS accumulation, and release of damage-associated molecular patterns, such as HMGB1 – is a form of immunogenic cell death that induces inflammatory cytokine secretion, PD-L1 upregulation on tumor cells, and functional maturation of dendritic cells in vitro. Intratumoral injection of P.a reprogrammed TMEs, increased CD8+ T cell infiltration, and led to synergistic tumor regression, without systemic toxicity when combined with anti-PD-L1 in an MC38 tumor model.

Contributed by Shishir Pant

Hu et al. demonstrated that Pseudomonas aeruginosa (P.a) initiates caspase-3-dependent apoptosis in MC38 CRC cell lines. Pyroptosis – characterized by GSDME cleavage, intracellular ROS accumulation, and release of damage-associated molecular patterns, such as HMGB1 – is a form of immunogenic cell death that induces inflammatory cytokine secretion, PD-L1 upregulation on tumor cells, and functional maturation of dendritic cells in vitro. Intratumoral injection of P.a reprogrammed TMEs, increased CD8+ T cell infiltration, and led to synergistic tumor regression, without systemic toxicity when combined with anti-PD-L1 in an MC38 tumor model.

Contributed by Shishir Pant

ABSTRACT: Colorectal cancer (CRC) exhibits limited responsiveness to immune checkpoint inhibitors (ICIs), largely due to its immunosuppressive tumor microenvironment (TME) and poor baseline immunogenicity. Here, we report that Pseudomonas aeruginosa (P. aeruginosa) triggers caspase-3-dependent pyroptosis in murine CRC MC38 cells, characterized by GSDME cleavage, intracellular reactive oxygen species (ROS) accumulation, and the release of damage-associated molecular patterns (DAMPs). This form of immunogenic cell death promotes robust inflammatory cytokine secretion, upregulation of PD-L1 on tumor cells, and functional maturation of bone marrow-derived dendritic cells (BMDCs) in vitro. In vivo, intratumoral injection of P. aeruginosa leads to significant reprogramming of the TME, including increased expression of proinflammatory genes, DC maturation, and enhanced infiltration of CD8(+) T lymphocytes. Notably, combination therapy with P. aeruginosa and an anti-PD-L1 antibody results in synergistic tumor regression, markedly outperforming either monotherapy, without inducing detectable systemic toxicity. Together, our findings reveal that P. aeruginosa-induced pyroptosis serves as a potent immunogenic stimulus that reshapes the CRC tumor microenvironment and overcomes resistance to immune checkpoint blockade. This strategy represents a promising approach to enhance immunotherapy efficacy in poorly immunogenic solid tumors.

Author Info: (1) The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210032, China. (2) Department of Neurology, Xindu District People's Hospital of Chengdu, Chengdu, 610500,

Author Info: (1) The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210032, China. (2) Department of Neurology, Xindu District People's Hospital of Chengdu, Chengdu, 610500, China. (3) The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210032, China. (4) Department of Clinical Microbiology, School of Laboratory Medicine, Chengdu Medical College, Clinical IVD Joint Research Center of Chengdu Medical College-Maccura Biotechnology, Chengdu, 610500, China. Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-Origin Food, Chengdu Medical College, Chengdu, 610500, China. (5) The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210032, China. (6) The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210032, China. (7) Department of Clinical Microbiology, School of Laboratory Medicine, Chengdu Medical College, Clinical IVD Joint Research Center of Chengdu Medical College-Maccura Biotechnology, Chengdu, 610500, China. Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-Origin Food, Chengdu Medical College, Chengdu, 610500, China. (8) Department of Clinical Microbiology, School of Laboratory Medicine, Chengdu Medical College, Clinical IVD Joint Research Center of Chengdu Medical College-Maccura Biotechnology, Chengdu, 610500, China. Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-Origin Food, Chengdu Medical College, Chengdu, 610500, China. (9) Taishan Community Health Service Center, Jiangbei New District, Nanjing, 210032, China. (10) Department of Neurology, Xindu District People's Hospital of Chengdu, Chengdu, 610500, China. 501838695@qq.com. (11) Department of Clinical Microbiology, School of Laboratory Medicine, Chengdu Medical College, Clinical IVD Joint Research Center of Chengdu Medical College-Maccura Biotechnology, Chengdu, 610500, China. renke@cmc.edu.cn. Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-Origin Food, Chengdu Medical College, Chengdu, 610500, China. renke@cmc.edu.cn. (12) The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210032, China. 2608236988@qq.com.

CAR-T triggers TAM reeducation and adaptive anti-tumor response via TREM2 deficiency or CD40 agonist Spotlight 

Liu, Gao, Xi, et al. showed that TREM2⁺ TAMs drive GPC3-CAR-T resistance in hepatocellular carcinoma. Trem2 deletion synergized with CAR-T therapy to enhance effector functionality (with reduced exhaustion in endogenous tumor-specific T cells), metabolically reprogram TAMs toward an antitumor CXCL9hi/SPP1lo phenotype, and achieve durable control in an HCC tumor model. The dual intervention enhanced oxidative metabolism and suppressed glycolysis via AMPK and STAT1 signaling and PI3K–AKT–mTOR inhibition. CD40 agonism phenocopied Trem2 loss, with sotigalimab promoting human CD8⁺ T cell migration and CAR-T responses.

Contributed by Shishir Pant

Liu, Gao, Xi, et al. showed that TREM2⁺ TAMs drive GPC3-CAR-T resistance in hepatocellular carcinoma. Trem2 deletion synergized with CAR-T therapy to enhance effector functionality (with reduced exhaustion in endogenous tumor-specific T cells), metabolically reprogram TAMs toward an antitumor CXCL9hi/SPP1lo phenotype, and achieve durable control in an HCC tumor model. The dual intervention enhanced oxidative metabolism and suppressed glycolysis via AMPK and STAT1 signaling and PI3K–AKT–mTOR inhibition. CD40 agonism phenocopied Trem2 loss, with sotigalimab promoting human CD8⁺ T cell migration and CAR-T responses.

Contributed by Shishir Pant

ABSTRACT: Chimeric antigen receptor (CAR)-T therapy targeting GPC3 shows unsatisfactory clinical efficacy in hepatocellular carcinoma (HCC). Combining clinical data and the immunocompetent orthotopic HCC model, we demonstrate that TREM2(+) tumor-associated macrophages (TAMs) are critical mediators of GPC3-CAR-T resistance. We find that Trem2 deficiency synergizes with GPC3-CAR-T to enhance tumor control by expanding endogenous tumor-specific CD8(+) T cells (not CAR-T amplification) and reeducating TAMs to an anti-tumor CXCL9(hi)/SPP1(lo) phenotype via metabolic reprogramming. Mechanistically, this combination enhances oxidative metabolism while suppressing glycolysis through JAK-STAT1 triggering, AMPK activation, and PI3K-AKT-mTOR inhibition. Crucially, Trem2 deficiency up-regulates CD40 expression, enabling CD40 agonism to phenocopy Trem2-deficiency effects via AMPK activation and STAT1-driven CXCL9 production. Notably, the clinical agonist sotigalimab similarly enhances human CD8(+) T cell migration in vitro. Our findings highlight the significance of combining GPC3-CAR-T therapy with CD40 agonist as a critical pre-requisite for eliciting reeducation of TAMs and enhancing the efficacy of CAR-T therapy in HCC.

Author Info: (1) The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Clinical Laboratory/State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Prov

Author Info: (1) The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Clinical Laboratory/State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China. (2) Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510600, China. (3) School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China; Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China. (4) The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China. (5) The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Clinical Laboratory/State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China. (6) The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Clinical Laboratory/State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China. (7) The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Clinical Laboratory/State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China. (8) The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Clinical Laboratory/State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China. (9) The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China. (10) The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Clinical Laboratory/State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China. (11) The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Clinical Laboratory/State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China. Electronic address: huangxz020@163.com. (12) The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China. Electronic address: fwzhe@mail.sysu.edu.cn. (13) The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China. Electronic address: tanjzh@mail.sysu.edu.cn.

Pan-cancer N-glycoproteomic atlas of patient-derived xenografts uncovers FAT2 as an actionable surface target

Cell surface proteins offer significant cancer therapeutic potential attributable to their accessible membrane localization and central roles in cellular signaling, yet their promise remains largely untapped due to technical challenges inherent to profiling them. Here, we employ N-glycoproteomics to analyze 85 patient-derived xenografts (PDXs), constructing Glyco PDXplorer-an in vivo pan-cancer atlas of cancer-derived surface proteins. We develop a target discovery pipeline to prioritize proteins with favorable expression profiles for immunotherapeutic targeting and validate FAT2 as a squamous-cancer-enriched surface protein minimally detected in normal tissue. Functional studies reveal that FAT2 is essential for head and neck squamous cancer (HNSC) cell growth and adhesion through regulation of surface architecture and integrin-PI3K signaling. Chimeric antigen receptor (CAR)-T cells targeting FAT2 demonstrate anti-tumor activity. This work lays the foundation for developing FAT2-targeted therapies and represents a pivotal platform to inform therapeutic target discovery across cancers.

Author Info: (1) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (

Author Info: (1) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (2) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (3) Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 1C7, Canada. (4) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (5) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (6) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (7) Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, Ottawa, ON K1A 0R6, Canada. (8) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (9) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (10) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (11) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (12) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (13) Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, Ottawa, ON K1A 0R6, Canada. (14) Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada. (15) Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 1C7, Canada. (16) Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 1C7, Canada. (17) Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada. (18) Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 1C7, Canada. (19) Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada. (20) Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 1C7, Canada. (21) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (22) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (23) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (24) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (25) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (26) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (27) adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada. (28) adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada. (29) adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada. (30) adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada. (31) adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada. (32) adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada. (33) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (34) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (35) Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada. (36) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada. (37) Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Otolaryngology-Head & Neck Surgery, University of Toronto, Toronto, ON M5S 3H2, Canada. (38) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (39) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. (40) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada. (41) adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada. (42) adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada. (43) Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 1C7, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada. (44) Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada. Electronic address: thomas.kislinger@utoronto.ca.

Optimally engineered HLA/peptide-specific CAR-T cells outperform TCR-T cells to eradicate solid tumors

Tumor-specific HLA/peptides (pHLA) represent attractive therapeutic targets for cancer. Two cell-based modalities can target pHLA-expressing tumors: T cell receptors (TCRs) or TCR-mimetic (TCRm) antibodies reformatted as chimeric antigen receptors (CARs). Using HLA-A2/MAGEA4(230-239) as a model pHLA, we discerned the relative potency of TCR-T and CAR-T cells, informing how to best deploy these for clinical benefit. Although TCR-T cells were more sensitive at detecting low-density pHLA, TCR-T cells exerted only transient in vivo antitumor efficacy followed by tumor relapse due to deficient TCR-T cell proliferation and persistence that was associated with a more differentiated and dysfunctional phenotype. By contrast, CAR-T cells with encoded costimulatory signaling fully regressed tumors. Insufficient TCR-T cell durability was overcome by coengaging 41BB or IL-2 signaling pathways, thereby enhancing tumor control in vivo. These data establish differential activities of human TCR-T and CAR-T cells targeting the same pHLA and inform the development of optimal targeting strategies to induce durable clinical responses.

Author Info: (1) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (2) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (3) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (4) Regenero

Author Info: (1) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (2) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (3) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (4) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (5) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (6) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (7) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (8) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (9) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (10) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (11) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (12) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (13) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (14) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (15) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (16) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA. (17) Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA.

Armored macrophage-targeted CAR-T cells reset and reprogram the tumor microenvironment and control metastatic cancer growth Featured  

Mateus-Tique et al. developed a CAR-T targeting FOLR2-expressing immunosuppressive tumor-associated macrophages (TAMs) with an IL-12 payload for local delivery. While high doses were toxic in a murine ovarian cancer model, injection with a low dose without lymphodepletion was tolerated and led to remodeling of the TME, including depletion of immunosuppressive TAMs and an increase in immune-stimulatory macrophages, resulting in reduced tumor growth and improved survival. Treatment induced endogenous antitumor CD8+ T cell responses, and the therapeutic effect was partially dependent on FAS. Targeting TREM2 with the same CAR-T strategy in a lung cancer model had similar effects.

Mateus-Tique et al. developed a CAR-T targeting FOLR2-expressing immunosuppressive tumor-associated macrophages (TAMs) with an IL-12 payload for local delivery. While high doses were toxic in a murine ovarian cancer model, injection with a low dose without lymphodepletion was tolerated and led to remodeling of the TME, including depletion of immunosuppressive TAMs and an increase in immune-stimulatory macrophages, resulting in reduced tumor growth and improved survival. Treatment induced endogenous antitumor CD8+ T cell responses, and the therapeutic effect was partially dependent on FAS. Targeting TREM2 with the same CAR-T strategy in a lung cancer model had similar effects.

ABSTRACT: Tumor-associated macrophages (TAMs), which commonly express FOLR2 or TREM2, are enriched in solid tumors and keep the tumor microenvironment (TME) immunosuppressed. Here, we introduce IL-12-expressing CAR-T cells targeting FOLR2 or TREM2 to deplete pro-tumor TAMs and reprogram the TME. Treatment with IL-12-armored anti-TAM CAR-T leads to significantly improved survival in metastatic ovarian and lung cancer models. The CAR-T mediates benefit at low cell dose and without lymphodepletion, and remains largely restricted to tumors with no overt toxicity. Spatial transcriptomics reveals that IL-12 anti-TAM CAR-T mediates sustained remodeling of the TME, even after CAR-T contraction, with the expansion of CXCL9+ immunostimulatory macrophages and endogenous tumor-specific cytotoxic T cells. Tumor clearance depends, in part, on FAS expression on cancer cells, revealing an IL-12-FAS axis for IL-12-armored CAR-T activity. These findings position IL-12-producing, myeloid-directed CAR-T as a broad strategy to remodel the TME and drive anti-tumor immunity for solid cancers.

Author Info: (1) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA

Author Info: (1) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (2) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (3) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (4) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (5) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (6) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (7) Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (8) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (9) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (10) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (11) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (12) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (13) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (14) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (15) Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (16) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (17) Department of Pathology and Immunology, Washington University, St Louis, MO, USA. (18) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (19) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (20) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. Electronic address: miriam.merad@mssm.edu. (21) Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. Electronic address: brian.brown@mssm.edu.

IGSF11-VISTA is a critical and targetable immune checkpoint axis in diffuse midline glioma Spotlight 

Using a multiomics approach on human and murine diffuse midline glioma samples, Collot, Ruiz-Moreno, Honhoff, et al. identified an MES pattern with mesenchymal tumor cells and blood-derived immune cells, and an AOO-pattern enriched with astrocyte, oligodendrocyte, and oligodendrocyte precursor-like cancer cell populations, alongside homeostatic-like microglia. Cancer cells in AOO niches expressed high levels of IGSF11 that signaled through VISTA on microglia. Targeting IGSF11–VISTA resulted in microglia-dependent, T cell-independent tumor reduction and survival benefit.

Contributed by Shishir Pant

Using a multiomics approach on human and murine diffuse midline glioma samples, Collot, Ruiz-Moreno, Honhoff, et al. identified an MES pattern with mesenchymal tumor cells and blood-derived immune cells, and an AOO-pattern enriched with astrocyte, oligodendrocyte, and oligodendrocyte precursor-like cancer cell populations, alongside homeostatic-like microglia. Cancer cells in AOO niches expressed high levels of IGSF11 that signaled through VISTA on microglia. Targeting IGSF11–VISTA resulted in microglia-dependent, T cell-independent tumor reduction and survival benefit.

Contributed by Shishir Pant

ABSTRACT: Diffuse midline glioma (DMG) is an aggressive pediatric brain tumor with no curative treatment, and lacks a comprehensive understanding of immune-tumor cell interactions within their spatial context. Our multi-omics approach, integrating single-nuclei RNA sequencing, spatial transcriptomics, and high-dimensional imaging, utilizes patient samples and an experimental murine DMG model to unveil two spatially distinct regions. MES-patterns are defined by mesenchymal (MES) tumor cells and blood-derived immune cells, whereas AOO-patterns are enriched with astrocyte (AC)-, oligodendrocyte (OC)-, and oligodendrocyte precursor cell (OPC)-like cancer populations, alongside homeostatic-like microglia. The less-studied immune checkpoint, IGSF11, is primarily expressed by AOO-associated cancer cells, while its receptor VISTA is detected mainly in homeostatic microglia. Targeting IGSF11-VISTA results in tumor reduction and survival benefit, mediated by brain-resident microglia and independent of T cell infiltration. This positions IGSF11-VISTA as a promising immune checkpoint treatment axis to harness the local brain immune response against DMG.

Author Info: (1) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (2) Princess M‡xima Center for Pediatric Oncology, Utrecht,

Author Info: (1) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (2) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, the Netherlands. (3) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (4) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (5) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (6) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (7) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (8) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (9) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (10) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands. (11) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands. (12) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (13) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (14) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (15) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands. (16) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (17) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (18) Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands. (19) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; University Medical Center Utrecht, Utrecht, the Netherlands; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Research Consortium (DKTK), Heidelberg, Germany. (20) Department of Bioengineering, Stanford University Schools of Engineering and Medicine, Stanford, CA, USA. (21) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands. (22) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands. (23) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands. (24) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (25) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands. (26) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, the Netherlands. (27) Princess M‡xima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands. Electronic address: a.c.rios@prinsesmaximacentrum.nl.

Optimal murine CD4+ T cell priming by mRNA-lipid nanoparticle vaccines requires endogenous antigen processing

Lipid nanoparticle (LNP)-encapsulated nucleoside-modified mRNA vaccines elicit robust CD4(+) T cell responses, yet the mechanisms underlying this T cell priming remain unknown. Antigens presented to CD4(+) T cells on major histocompatibility complex class II (MHC II) are traditionally acquired by antigen presenting cells (APCs) from extracellular sources. Here we show that vaccine-specific CD4(+) T cell responses instead rely on antigen directly expressed within APCs, without extracellular transit. Murine APCs treated with mRNA-LNP vaccines activate T cells more efficiently when presenting antigen produced internally, rather than acquired externally. Immunization with mRNA-LNP vaccines engineered to inhibit antigen expression in APCs results in lower antigen-specific CD4(+) T cell, T follicular helper cell, and antibody responses in mice. In contrast, excluding vaccine antigen from muscle cells minimally affects CD4(+) T cell responses. Our findings demonstrate that endogenous antigen presentation is essential to mRNA-LNP vaccine-induced immune responses and refine paradigms of MHC II-restricted antigen processing and presentation.

Author Info: (1) Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA. ROODJ@chop.edu. (2) School of Engineering and Applied Science, University of Pennsylvania,

Author Info: (1) Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA. ROODJ@chop.edu. (2) School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA. Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. (3) Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA. Bristol Myers Squibb, Lawrenceville, NJ, USA. (4) Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. (5) Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA. Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA. (6) Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA. Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA. (7) Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA. Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA. (8) Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA. Drexel University College of Medicine at Tower Health, Wyomissing, PA, USA. (9) Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. (10) Genevant Sciences Corporation, Vancouver, British Columbia, Canada. (11) Genevant Sciences Corporation, Vancouver, British Columbia, Canada. (12) Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. (13) Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. (14) Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA. (15) Genevant Sciences Corporation, Vancouver, British Columbia, Canada. (16) Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. (17) Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA. eisenlc@pennmedicine.upenn.edu. Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. eisenlc@pennmedicine.upenn.edu.

Single-cell screens identify ADAM12 as a fibroblast checkpoint impeding anti-tumor immunity Spotlight 

Li, Liu, Guo, and Zhang et al. employed a dual CRISPR-inhibitor and -activator functional Perturb-seq single-cell screening, coupled with in silico genomic analysis of patient-derived tumor and tumor-adjacent fibroblasts. An antagonistic relationship was identified between a pro-tumoral TGFβ-dependent myofibroblast program and an antitumoral IFN-I responsive program, which was mediated by ADAM12. Loss of fibroblast-specific ADAM12 enhanced IFN-I signaling and restricted TGFβ signaling, resulting in TME remodeling, enhanced CD8+ T cell numbers and function, and decreased tumor burden in multiple models, boostable by ICB.

Contributed by Katherine Turner

Li, Liu, Guo, and Zhang et al. employed a dual CRISPR-inhibitor and -activator functional Perturb-seq single-cell screening, coupled with in silico genomic analysis of patient-derived tumor and tumor-adjacent fibroblasts. An antagonistic relationship was identified between a pro-tumoral TGFβ-dependent myofibroblast program and an antitumoral IFN-I responsive program, which was mediated by ADAM12. Loss of fibroblast-specific ADAM12 enhanced IFN-I signaling and restricted TGFβ signaling, resulting in TME remodeling, enhanced CD8+ T cell numbers and function, and decreased tumor burden in multiple models, boostable by ICB.

Contributed by Katherine Turner

ABSTRACT: Clinical trials targeting cancer-associated fibroblasts (CAFs)-crucial pro-tumoral factors in cancer-have almost all failed. This may be ascribed to their intrinsic functional plasticity and the opaque regulatory circuits underlying their heterogeneous phenotypes within tumors. We address these by developing a systematic screening approach for patient-derived fibroblasts using complementary CRISPR interference (CRISPRi) and activation (CRISPRa)-based Perturb-seq. An anti-tumoral interferon (IFN)-I response-associated program is identified as the primary antagonism axis counteracting TGF-β-driven pro-tumoral myofibroblast activation. ADAM12 emerges as a molecular checkpoint mediating this relationship. Its ablation elicits IFN-I-responsive programs, reconfigures myofibroblast population structures into progenitor-like states, revitalizes T cell-based immune responses, and induces tumor rejection across various murine models. Further combined with human genomics data analysis, our findings position ADAM12 as a potential target for fibroblasts, paving the way for actionable therapeutic interventions.

Author Info: (1) Changping Laboratory, Beijing 102206, China; Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National

Author Info: (1) Changping Laboratory, Beijing 102206, China; Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China; Peking University Beijing-Tianjin-Hebei Biomedical Pioneering Innovation Center, Tianjin 300405, China. Electronic address: lijianan@pku.edu.cn. (2) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (3) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China; Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China. (4) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (5) State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. (6) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (7) Analytical Biosciences Limited, Beijing 100084, China. (8) Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China. (9) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China; Institute for Data-Driven Tumor Immunology, Chongqing Medical University, Chongqing 400016, China. (10) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (11) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (12) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (13) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (14) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (15) Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China. (16) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (17) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (18) GeneX Health Co, Led, Beijing 100195, China. (19) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (20) Analytical Biosciences Limited, Beijing 100084, China; Peking University Beijing-Tianjin-Hebei Biomedical Pioneering Innovation Center, Tianjin 300405, China. (21) Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Ninth School of Clinical Medicine, Peking University, Beijing 100038, China. (22) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. (23) State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. Electronic address: jzx@pku.edu.cn. (24) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China. Electronic address: wangdf19@pku.edu.cn. (25) Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Academy for Advanced Interdisciplinary Studies, National Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention and Treatment, Peking University, Beijing 100871, China; Institute for Data-Driven Tumor Immunology, Chongqing Medical University, Chongqing 400016, China. Electronic address: zemin@pku.edu.cn.

ROSE12, a novel anti-CTLA-4 FcγRs binding-enhanced antibody activated by extracellular adenosine triphosphate, shows tumor-selective regulatory T-cell depletion and antitumor efficacy without systemic immune activation

Spotlight 

To improve safety for Treg-depleting immunotherapy, Hayashi, Tatsumi, Katada, Matsuda, et al. generated ROSE12, a novel ATP-activated anti-CTLA-4 “switch” mAb with enhanced binding to FcγRIIa and FcγRIIIa, and decreased binding to FcγRIIb. As extracellular ATP levels are 1000-fold higher in the TME compared to normal tissues, ROSE12 showed superior tumor-selective Treg reduction (via ADCC and ADCP) and increased CD8+ T cell infiltration, with no systemic immune activation. In mouse models, ROSE12 significantly inhibited tumor growth and showed synergistic efficacy with anti-PD-L1, and is currently in phase I clinical trials.

Contributed by Katherine Turner

To improve safety for Treg-depleting immunotherapy, Hayashi, Tatsumi, Katada, Matsuda, et al. generated ROSE12, a novel ATP-activated anti-CTLA-4 “switch” mAb with enhanced binding to FcγRIIa and FcγRIIIa, and decreased binding to FcγRIIb. As extracellular ATP levels are 1000-fold higher in the TME compared to normal tissues, ROSE12 showed superior tumor-selective Treg reduction (via ADCC and ADCP) and increased CD8+ T cell infiltration, with no systemic immune activation. In mouse models, ROSE12 significantly inhibited tumor growth and showed synergistic efficacy with anti-PD-L1, and is currently in phase I clinical trials.

Contributed by Katherine Turner

Background: Intratumoral regulatory T cells (Tregs) are associated with diminished antitumor immunity and poor prognosis in many cancers, with tumor-infiltrating effector Tregs expressing high levels of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). While Treg depletion is a promising strategy for cancer immunotherapy, systemic Treg depletion may lead to severe autoimmune toxicity. Therefore, to selectively deplete intratumoral Tregs, we used extracellular ATP (exATP), which is highly elevated in solid tumors, as a tumor-selective small molecule.

Methods: We generated ROSE12, a novel anti-CTLA-4 Fc gamma receptors (FcγRs)-binding-enhanced-Fc exATP-dependent switch antibody that reduces Tregs only in the presence of exATP. We evaluated ATP-dependent binding affinity, antibody-dependent cellular cytotoxicity (ADCC) activity in vitro, and antitumor efficacy of monotherapy and combination therapy with anti-programmed death-ligand 1 (PD-L1) in CTLA-4/CD3 double humanized mouse models. Safety profiles were assessed in cynomolgus monkeys.

Results: ROSE12 demonstrated ATP concentration-dependent binding to CTLA-4, with strong binding at 100 µmol/L but no binding without ATP. ROSE12 demonstrated stronger exATP-dependent ADCC activity in vitro and preferentially reduced CTLA-4+ Tregs over activated conventional T cells. The engineered asymmetric re-engineering technology-Fc (ART-Fc) region, a proprietary Fc engineering technology, showed enhanced binding to activating FcγRIIa and FcγRIIIa while reducing binding to inhibitory FcγRIIb. In mouse models, ROSE12 monotherapy significantly inhibited tumor growth in both conventional and PD-L1 therapy-resistant tumors by reducing intratumoral Tregs and increasing CD8+ T-cell infiltration. Combination therapy with anti-PD-L1 showed synergistic antitumor efficacy with enhanced intratumoral CD8+ T-cell activation without increasing systemic immune activation. Unlike FcγRs binding-enhanced conventional anti-CTLA-4, ROSE12 did not induce systemic immune activation or colitis symptoms, demonstrating a 30-300-fold wider therapeutic window. The tumor-selective mechanism was confirmed in humanized mouse models, where ROSE12 reduced only intratumoral Tregs while sparing splenic Tregs. In cynomolgus monkeys, ROSE12 was well tolerated even at 30 mg/kg/week compared with the 3-10 mg/kg/week limits for conventional anti-CTLA-4 antibodies such as non-fucosylated ipilimumab and ipilimumab.

Conclusions: These findings support the clinical development of ROSE12 as a tumor-selective Treg-depleting immunotherapy with potential efficacy in programmed cell death protein-1/PD-L1 therapy-resistant patients. The favorable safety profile was attributed to the ATP-dependent binding mechanism that restricts activity to the high-ATP tumor microenvironment. ROSE12 is currently being evaluated in phase I clinical trials (NCT05907980).

Author Info: (1) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan hayashi.hiroki24@chugai-pharm.co.jp. (2) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanag

Author Info: (1) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan hayashi.hiroki24@chugai-pharm.co.jp. (2) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (3) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (4) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (5) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (6) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (7) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (8) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (9) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (10) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (11) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (12) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Chuo-ku, Tokyo, Japan. (13) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (14) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (15) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (16) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (17) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (18) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (19) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (20) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (21) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (22) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (23) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (24) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (25) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (26) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (27) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (28) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Yokohama, Kanagawa, Japan. (29) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (30) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (31) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (32) Chugai Pharmaceutical Co., Ltd., Translational Research Div, Chuo-ku, Tokyo, Japan. (33) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (34) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (35) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (36) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (37) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (38) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (39) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (40) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (41) Experimental Immunology, Immunology Frontier Research Center; Osaka University, Suita, Osaka, Japan. (42) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan. (43) Chugai Pharmaceutical Co., Ltd., Research Div, Yokohama, Kanagawa, Japan.

Extracellular vesicles are key mediators for direct antigen transport to draining lymph nodes Spotlight 

Wang and Cheng et al. showed that electrotransfection activated the HSP90–p53–TSAP6 signaling pathway to enhance extracellular vesicle (EV) biogenesis, facilitating antigen transport from muscles to draining lymph nodes during DNA vaccination. Vaccine-encoded transmembrane antigen (hemagglutinin) was enriched in the secreted EVs, trafficked through lymphatic vessels, and reached draining lymph nodes within hours. Direct visualization confirmed passive extracellular vesicle transport through lymphatic vasculature. Pharmacologic inhibition of vesicle biogenesis reduced EV concentration in the muscle, and impaired immune responses.

Contributed by Shishir Pant

Wang and Cheng et al. showed that electrotransfection activated the HSP90–p53–TSAP6 signaling pathway to enhance extracellular vesicle (EV) biogenesis, facilitating antigen transport from muscles to draining lymph nodes during DNA vaccination. Vaccine-encoded transmembrane antigen (hemagglutinin) was enriched in the secreted EVs, trafficked through lymphatic vessels, and reached draining lymph nodes within hours. Direct visualization confirmed passive extracellular vesicle transport through lymphatic vasculature. Pharmacologic inhibition of vesicle biogenesis reduced EV concentration in the muscle, and impaired immune responses.

Contributed by Shishir Pant

ABSTRACT: DNA vaccines have shown great potential in preclinical and clinical studies. However, it is still unclear how the antigen expressed at the site of vaccination is delivered to draining lymph nodes for activation of the immune system. To address the issue, the current study investigated the role of extracellular vesicles (EVs) in the delivery. Following intramuscular electrotransfection of DNA vaccines encoding a transmembrane antigen, hemagglutinin (HA), EV secretion was significantly increased in the muscle with the peak level being ∼10-fold higher than the unvaccinated control. More importantly, the EVs were highly enriched with HA, and could reach the draining lymph nodes through lymphatic vessels within 4 h. Blocking the EV secretion by systemic treatment with a small molecular inhibitor, GW4869, significantly reduced humoral and cellular responses against the antigen. These findings indicated that the EVs play an important role in the antigen delivery, suggesting that enhancing local EV biogenesis and antigen packaging into EVs can be new avenues for development of next-generation vaccine adjuvants.

Author Info: (1) Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA. (2) Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA. (3) Depart

Author Info: (1) Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA. (2) Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA. (3) Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA. Electronic address: ke.cheng@columbia.edu. (4) Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA. Electronic address: fyuan@duke.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.