Journal Articles

Programming tissue-sensing T cells that deliver therapies to the brain Featured  

Two teams of researchers working with lead author Dr. Wendell Lim demonstrated the use of synNotch-engineered CD4+ T cells directed towards tissue-specific antigens, triggering the local release of various disease-specific payloads. Reddy et al. utilized this technology to suppress CAR T cell cytotoxicity in on-target/off-tumor tissues and to suppress immune rejection in transplanted organs, while Simic and Watchmaker et al. demonstrated its ability to induce CAR expression only in the brain and to specifically deliver payloads that suppress neuroinflammation. 

Also review: Engineering synthetic suppressor T cells that execute locally targeted immunoprotective programs

Two teams of researchers working with lead author Dr. Wendell Lim demonstrated the use of synNotch-engineered CD4+ T cells directed towards tissue-specific antigens, triggering the local release of various disease-specific payloads. Reddy et al. utilized this technology to suppress CAR T cell cytotoxicity in on-target/off-tumor tissues and to suppress immune rejection in transplanted organs, while Simic and Watchmaker et al. demonstrated its ability to induce CAR expression only in the brain and to specifically deliver payloads that suppress neuroinflammation. 

Also review: Engineering synthetic suppressor T cells that execute locally targeted immunoprotective programs

ABSTRACT: To engineer cells that can specifically target the central nervous system (CNS), we identified extracellular CNS-specific antigens, including components of the CNS extracellular matrix and surface molecules expressed on neurons or glial cells. Synthetic Notch receptors engineered to detect these antigens were used to program T cells to induce the expression of diverse payloads only in the brain. CNS-targeted T cells that induced chimeric antigen receptor expression efficiently cleared primary and secondary brain tumors without harming cross-reactive cells outside of the brain. Conversely, CNS-targeted cells that locally delivered the immunosuppressive cytokine interleukin-10 ameliorated symptoms in a mouse model of neuroinflammation. Tissue-sensing cells represent a strategy for addressing diverse disorders in an anatomically targeted manner.

Author Info: (1) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. (2) Department of Neurological S

Author Info: (1) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. (2) Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA. (3) Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA. (4) Department of Computer Science, Princeton University, Princeton, NJ, USA. Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA. (5) Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA. (6) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. (7) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. (8) Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA. (9) Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA. (10) Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA. (11) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. (12) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. (13) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. (14) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. (15) School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada. (16) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. (17) School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada. (18) Department of Computer Science, Princeton University, Princeton, NJ, USA. Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA. Center for Computational Biology, Flatiron Institute, New York, NY, USA. (19) Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA. (20) Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA. (21) Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA. Program in Immunology, University of California San Francisco, San Francisco, CA, USA. (22) Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA. Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA. Helen Diller Cancer Center, University of California San Francisco, San Francisco, CA, USA. (23) UCSF Cell Design Institute and Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA. Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.

Engineering synthetic suppressor T cells that execute locally targeted immunoprotective programs Featured  

Two teams of researchers working with lead author Dr. Wendell Lim demonstrated the use of synNotch-engineered CD4+ T cells directed towards tissue-specific antigens, triggering the local release of various disease-specific payloads. Reddy et al. utilized this technology to suppress CAR T cell cytotoxicity in on-target/off-tumor tissues and to suppress immune rejection in transplanted organs, while Simic and Watchmaker et al. demonstrated its ability to induce CAR expression only in the brain and to specifically deliver payloads that suppress neuroinflammation. 

Also review: Programming tissue-sensing T cells that deliver therapies to the brain

Two teams of researchers working with lead author Dr. Wendell Lim demonstrated the use of synNotch-engineered CD4+ T cells directed towards tissue-specific antigens, triggering the local release of various disease-specific payloads. Reddy et al. utilized this technology to suppress CAR T cell cytotoxicity in on-target/off-tumor tissues and to suppress immune rejection in transplanted organs, while Simic and Watchmaker et al. demonstrated its ability to induce CAR expression only in the brain and to specifically deliver payloads that suppress neuroinflammation. 

Also review: Programming tissue-sensing T cells that deliver therapies to the brain

ABSTRACT: Immune homeostasis requires a balance of inflammatory and suppressive activities. To design cells potentially useful for local immune suppression, we engineered conventional CD4(+) T cells with synthetic Notch (synNotch) receptors driving antigen-triggered production of anti-inflammatory payloads. Screening a diverse library of suppression programs, we observed the strongest suppression of cytotoxic T cell attack by the production of both anti-inflammatory factors (interleukin-10, transforming growth factor-_1, programmed death ligand 1) and sinks for proinflammatory cytokines (interleukin-2 receptor subunit CD25). Engineered cells with bespoke regulatory programs protected tissues from immune attack without systemic suppression. Synthetic suppressor T cells protected transplanted beta cell organoids from cytotoxic T cells. They also protected specific tissues from unwanted chimeric antigen receptor (CAR) T cell cross-reaction. Synthetic suppressor T cells are a customizable platform to potentially treat autoimmune diseases, organ rejection, and CAR T cell toxicities with spatial precision.

Author Info: (1) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Cellular and Molecular Pharmacology, University of California, San Fr

Author Info: (1) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA. (2) Diabetes Center, University of California, San Francisco, San Francisco, CA, USA. (3) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA. Diabetes Center, University of California, San Francisco, San Francisco, CA, USA. (4) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA. (5) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA. (6) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA. (7) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA. (8) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA. (9) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA. (10) UCSF CoLabs, University of California, San Francisco, San Francisco, CA, USA. (11) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA. Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA. Department of Medicine, University of California San Francisco, San Francisco, CA, USA. (12) Diabetes Center, University of California, San Francisco, San Francisco, CA, USA. (13) Diabetes Center, University of California, San Francisco, San Francisco, CA, USA. (14) UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA. Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA. Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.

Chemotherapy induces myeloid-driven spatially confined T cell exhaustion in ovarian cancer Spotlight 

Launonen et al. explored the dynamic remodeling of HGSC TME under platinum-based chemotherapy and identified myelonets – spatially confined networks of interconnected myeloid cells – as key drivers of T cell exhaustion. Chemotherapy induced T cell infiltration and exhaustion, particularly within myeloid-rich areas. At the tumor–stroma interface, M2 macrophages excluded T cell interaction with tumor cells, while M1 macrophages at myelonets induced exhaustion through NECTIN2–TIGIT signaling. Targeting the chemotherapy-induced TIGIT–NECTIN2 axis in patient-derived 3D cultures reactivated T cells, highlighting a therapeutic opportunity.

Contributed by Shishir Pant

Launonen et al. explored the dynamic remodeling of HGSC TME under platinum-based chemotherapy and identified myelonets – spatially confined networks of interconnected myeloid cells – as key drivers of T cell exhaustion. Chemotherapy induced T cell infiltration and exhaustion, particularly within myeloid-rich areas. At the tumor–stroma interface, M2 macrophages excluded T cell interaction with tumor cells, while M1 macrophages at myelonets induced exhaustion through NECTIN2–TIGIT signaling. Targeting the chemotherapy-induced TIGIT–NECTIN2 axis in patient-derived 3D cultures reactivated T cells, highlighting a therapeutic opportunity.

Contributed by Shishir Pant

ABSTRACT: Anti-tumor immunity is crucial for high-grade serous ovarian cancer (HGSC) prognosis, yet its adaptation upon standard chemotherapy remains poorly understood. Here, we conduct spatial and molecular characterization of 117 HGSC samples collected before and after chemotherapy. Our single-cell and spatial analyses reveal increasingly versatile immune cell states forming spatiotemporally dynamic microcommunities. We describe Myelonets, networks of interconnected myeloid cells that contribute to CD8(+) T cell exhaustion post-chemotherapy and show that M1/M2 polarization at the tumor-stroma interface is associated with CD8(+) T cell exhaustion and exclusion, correlating with poor chemoresponse. Single-cell and spatial transcriptomics reveal prominent myeloid-T cell interactions via NECTIN2-TIGIT induced by chemotherapy. Targeting these interactions using a functional patient-derived immuno-oncology platform demonstrates that high NECTIN2-TIGIT signaling in matched tumors predicts responses to immune checkpoint blockade. Our discovery of clinically relevant myeloid-driven spatial T cell exhaustion unlocks immunotherapeutic strategies to unleash CD8(+) T cell-mediated anti-tumor immunity in HGSC.

Author Info: (1) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (2) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (3) Resear

Author Info: (1) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (2) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (3) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (4) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (5) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (6) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (7) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (8) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (9) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (10) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (11) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (12) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (13) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (14) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (15) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (16) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Obstetrics and Gynecology, Department of Oncology, Clinical Trials Unit, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland. (17) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland. (18) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (19) Department of Obstetrics and Gynecology, University of Turku and Turku University Hospital, Turku, Finland. (20) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland. (21) Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA. (22) Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA; Ludwig Center at Harvard, Boston, MA, USA. (23) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland. (24) Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA; Ludwig Center at Harvard, Boston, MA, USA. (25) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Foundation for the Finnish Cancer Institute, Helsinki, Finland. Electronic address: anna.vaharautio@helsinki.fi. (26) Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Obstetrics and Gynecology, Department of Oncology, Clinical Trials Unit, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland; Institute for Molecular Medicine Finland, Helsinki Institute for Life Sciences, University of Helsinki, Helsinki, Finland. Electronic address: anniina.farkkila@helsinki.fi.

Intratumoral delivery of lipid nanoparticle-formulated mRNA encoding IL-21, IL-7, and 4-1BBL induces systemic anti-tumor immunity Spotlight 

Hamouda et al. engineered LNPs with an ionizable cationic lipid S-Ac7-DOG to improve levels of cargo expression and reduce reactogenicity. Intratumoral (i.t.) delivery of such LNPs, comprising a trio of mRNAs encoding IL−21, IL-7, and 4-1BBL (Triplet LNP), led to efficient expression of the mediators in tumor and myeloid cells. In multiple murine tumor models, Triplet LNP i.t. delivery immunologically activated the TME and TDLNs, induced systemic immunity to provoke CD8+ T cell-mediated therapeutic efficacy against directly treated and distal tumors, supported memory responses upon tumor rechallenge, and synergized with anti-PD-1, even in ICB-resistant models.

Contributed by Paula Hochman

Hamouda et al. engineered LNPs with an ionizable cationic lipid S-Ac7-DOG to improve levels of cargo expression and reduce reactogenicity. Intratumoral (i.t.) delivery of such LNPs, comprising a trio of mRNAs encoding IL−21, IL-7, and 4-1BBL (Triplet LNP), led to efficient expression of the mediators in tumor and myeloid cells. In multiple murine tumor models, Triplet LNP i.t. delivery immunologically activated the TME and TDLNs, induced systemic immunity to provoke CD8+ T cell-mediated therapeutic efficacy against directly treated and distal tumors, supported memory responses upon tumor rechallenge, and synergized with anti-PD-1, even in ICB-resistant models.

Contributed by Paula Hochman

ABSTRACT: Local delivery of mRNA-based immunotherapy offers a promising avenue as it enables the production of specific immunomodulatory proteins that can stimulate the immune system to recognize and eliminate cancer cells while limiting systemic exposure and toxicities. Here, we develop and employ lipid-based nanoparticles (LNPs) to intratumorally deliver an mRNA mixture encoding the cytokines interleukin (IL)-21 and IL-7 and the immunostimulatory molecule 4-1BB ligand (Triplet LNP). IL-21 synergy with IL-7 and 4-1BBL leads to a profound increase in the frequency of tumor-infiltrating CD8+ T cells and their capacity to produce granzyme B and IFN-γ, leading to tumor eradication and the development of long-term immunological memory. Mechanistically, the efficacy of the Triplet LNP depends on tumor-draining lymph nodes to tumor CD8+ T-cell trafficking. Moreover, we highlight the therapeutic potential of the Triplet LNP in multiple tumor models in female mice and its superior therapeutic efficacy to immune checkpoint blockade. Ultimately, the expression of these immunomodulators is associated with better overall survival in patients with cancer.

Author Info: (1) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for I

Author Info: (1) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (2) etherna, Ghent, Belgium. (3) etherna, Ghent, Belgium. (4) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (5) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (6) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (7) etherna, Ghent, Belgium. (8) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (9) Liver Cell Biology Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (10) Department of Anatomo-Pathology, Universitair Ziekenhuis Brussel (UZB), Brussels, Belgium. (11) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (12) Department of Pharmaceutics, University of Ghent, Ghent, Belgium. (13) etherna, Ghent, Belgium. (14) etherna, Ghent, Belgium. (15) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium. (16) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (17) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium. (18) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium. (19) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium. (20) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium. (21) CDL Research, University Medical Center, Utrecht, The Netherlands. (22) CDL Research, University Medical Center, Utrecht, The Netherlands. (23) Liver Cell Biology Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (24) Institut de Duve, UniversitŽ Catholique de Louvain, Brussels, Belgium. Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium. (25) Department of Pharmaceutics, University of Ghent, Ghent, Belgium. (26) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. (27) etherna, Ghent, Belgium. (28) etherna, Ghent, Belgium. florence.lambolez@etherna.be. (29) Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium. dlaoui@vub.be. Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium. dlaoui@vub.be.

Pyroptotic cell corpses are crowned with F-actin-rich filopodia that engage CLEC9A signaling in incoming dendritic cells Spotlight 

Holley et al. investigated the morphological changes during pyroptotic cell death, and identified that inflammasome activation induces F-actin-rich filopodia formation before plasma membrane rupture. Gasdermin D pores facilitate calcium influx, triggering actin polymerization and crowning of the resultant corpse with filopodia. Conventional dendritic cells recognize pyroptotic filopodia through the F-actin receptor, CLEC9A. This recognition triggered signaling via spleen tyrosine kinase (SYK), essential for antigen cross-presentation to T cells, promoting the transition from innate to adaptive immune response.

Contributed by Shishir Pant

Holley et al. investigated the morphological changes during pyroptotic cell death, and identified that inflammasome activation induces F-actin-rich filopodia formation before plasma membrane rupture. Gasdermin D pores facilitate calcium influx, triggering actin polymerization and crowning of the resultant corpse with filopodia. Conventional dendritic cells recognize pyroptotic filopodia through the F-actin receptor, CLEC9A. This recognition triggered signaling via spleen tyrosine kinase (SYK), essential for antigen cross-presentation to T cells, promoting the transition from innate to adaptive immune response.

Contributed by Shishir Pant

ABSTRACT: While apoptosis dismantles the cell to enforce immunological silence, pyroptotic cell death provokes inflammation. Little is known of the structural architecture of cells undergoing pyroptosis, and whether pyroptotic corpses are immunogenic. Here we report that inflammasomes trigger the Gasdermin-D- and calcium-dependent eruption of filopodia from the plasma membrane minutes before pyroptotic cell rupture, to crown the resultant corpse with filopodia. As a rich store of F-actin, pyroptotic filopodia are recognized by dendritic cells through the F-actin receptor, CLEC9A (DNGR1). We propose that cells assemble filopodia before cell rupture to serve as a posthumous mark for a cell that has died by gasdermin-induced pyroptosis, or MLKL-induced necroptosis, for recognition by dendritic cells. This study reveals the spectacular morphology of pyroptosis and identifies a mechanism by which inflammasomes induce pyroptotic cells to construct a de novo alarmin that activates dendritic cells via CLEC9A, which coordinates the transition from innate to adaptive immunity(1,2).

Author Info: (1) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. Max Planck Institute for Infection Biology, Berlin, Germany. (2) Institute for Mo

Author Info: (1) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. Max Planck Institute for Infection Biology, Berlin, Germany. (2) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. m.monteleone@imb.uq.edu.au. (3) Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA. (4) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia. (5) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia. (6) Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA. (7) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (8) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (9) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (10) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (11) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (12) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. Faculty of Science, University of Queensland, Brisbane, Queensland, Australia. (13) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (14) Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia. (15) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (16) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (17) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia. (18) Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA. (19) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (20) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. (21) Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia. K.Schroder@imb.uq.edu.au.

Regional immune mechanisms enhance efficacy of an autologous cellular cancer vaccine with intraperitoneal administration

Widespread peritoneal dissemination is common in patients with gynecologic or gastrointestinal cancers. Accumulating evidence of a central role for regional immunity in cancer control indicates that intraperitoneal immunotherapy may have treatment advantages. This study delineates immune mechanisms engaged by intraperitoneal delivery of a cell-based vaccine comprised of silicified ovarian cancer cells associated with enhanced survival. Vaccine trafficking from the site of injection to milky spots and other fat-associated lymphoid clusters was studied in syngeneic cancer models using bioluminescent and fluorescent imaging, microscopy, and flow cytometry. Spectral flow cytometry was used to phenotype peritoneal immune cell populations, while bioluminescent imaging of cancer was used to study myeloid and T cell dependency, systemic immunity, and vaccine efficacy in models of disseminated high-grade serous ovarian and DNA mismatch-repair proficient microsatellite-stable colorectal cancer. Following intraperitoneal vaccination of mice with ovarian cancer, vaccine cells were rapidly internalized by myeloid cells, with subsequent trafficking to fat-associated lymphoid clusters. Tumor clearance was confirmed to be T cell-mediated, leading to the establishment of local and systemic immunity. Combination immune checkpoint inhibitor and vaccine therapy in mice with advanced disease, characterized by an established suppressive tumor microenvironment, increased the number of mice with non-detectable tumors, however, change in tumor burden compared to vaccine monotherapy was not significant. Vaccination also resulted in tumor clearance in mouse models of metastatic colorectal cancer. This study demonstrates that intraperitoneal vaccine delivery has the potential to enhance vaccine efficacy by activating resident immune cells with the subsequent establishment of protective systemic anti-tumor immunity.

Author Info: (1) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA. (2) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM,

Author Info: (1) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA. (2) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA. (3) Department of Obstetrics & Gynecology, University of New Mexico Health Science Center, Albuquerque, NM, USA. (4) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA. (5) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA. (6) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA. (7) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA. (8) Department of Obstetrics & Gynecology, University of New Mexico Health Science Center, Albuquerque, NM, USA. (9) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA. (10) Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA.

High-resolution profile of neoantigen-specific TCR activation links moderate stimulation to increased resilience of engineered TCR-T cells

Neoantigen-specific T cell receptors (neoTCRs) promise safe, personalized anti-tumor immunotherapy. However, detailed assessment of neoTCR-characteristics affecting therapeutic efficacy is mostly missing. Previously, we identified diverse neoTCRs restricted to different neoantigens in a melanoma patient. In this work, we now combine single-cell TCR-sequencing and RNA-sequencing after neoantigen-specific restimulation of peripheral blood-derived CD8(+) T cells of this patient. We detect neoTCRs with specificity for the previously detected neoantigens and perform fine-characterization of neoTCR-transgenic (tg) T cells in vitro and in vivo. We describe a heterogeneous spectrum of TCR-intrinsic activation patterns in response to a shared neoepitope ranging from previously detected more highly frequent neoTCRs with moderate activation to rare ones with initially stronger activation. Experimental restimulation of adoptively transferred neoTCR-tg T cells in a xenogeneic rechallenge tumor model demonstrates superior anti-tumor responses of moderate neoTCR-tg T cells upon repeated tumor contact. These insights have significant implications for the selection of TCRs for therapeutic engineering of TCR-tg T cells.

Author Info: (1) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. (2) Technical Univer

Author Info: (1) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. (2) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. (3) Ludwig-Maximilians-UniversitŠt MŸnchen, Institute of Clinical Neuroimmunology, University Hospital, Marchioninistr. 15, 81377, Munich, Germany. Ludwig-Maximilians-UniversitŠt MŸnchen, Faculty of Medicine, Biomedical Center (BMC), Gro§haderner Str. 9, 82152, Martinsried, Germany. (4) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. (5) Technische UniversitŠt MŸnchen, Institute for Medical Microbiology, Immunology and Hygiene, Trogerstr. 30, 81675, Munich, Germany. (6) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. (7) Technische UniversitŠt MŸnchen, Institute for Medical Microbiology, Immunology and Hygiene, Trogerstr. 30, 81675, Munich, Germany. (8) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. (9) Technische UniversitŠt MŸnchen, Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine and Health, Ismaninger Str. 22, 81675, Munich, Germany. Technical University of Munich, Center for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Einsteinstr. 25, 81675, Munich, Germany. (10) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. (11) Technische UniversitŠt MŸnchen, Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine and Health, Ismaninger Str. 22, 81675, Munich, Germany. (12) Technical University of Munich, Center for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Einsteinstr. 25, 81675, Munich, Germany. Bavarian Center for Biomolecular Mass Spectrometry at Klinikum rechts der Isar, Technical University of Munich, Einsteinstr. 25, 81675, Munich, Germany. (13) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. German Cancer Consortium (DKTK), Partner-Site Munich and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. (14) Technische UniversitŠt MŸnchen, Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine and Health, Ismaninger Str. 22, 81675, Munich, Germany. Technical University of Munich, Center for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Einsteinstr. 25, 81675, Munich, Germany. (15) Technische UniversitŠt MŸnchen, Institute for Medical Microbiology, Immunology and Hygiene, Trogerstr. 30, 81675, Munich, Germany. German Center for Infection Research (Deutsches Zentrum fŸr Infektionsforschung, DZIF), Partner Site Munich, Munich, Germany. (16) Ludwig-Maximilians-UniversitŠt MŸnchen, Institute of Clinical Neuroimmunology, University Hospital, Marchioninistr. 15, 81377, Munich, Germany. Ludwig-Maximilians-UniversitŠt MŸnchen, Faculty of Medicine, Biomedical Center (BMC), Gro§haderner Str. 9, 82152, Martinsried, Germany. Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377, Munich, Germany. (17) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. German Cancer Consortium (DKTK), Partner-Site Munich and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. (18) Technical University of Munich, School of Medicine and Health, III Medical Department, TUM University Hospital, Ismaninger Str. 22, 81675, Munich, Germany. angela.krackhardt@tum.de. Technical University of Munich, Center for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Einsteinstr. 25, 81675, Munich, Germany. angela.krackhardt@tum.de. German Cancer Consortium (DKTK), Partner-Site Munich and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. angela.krackhardt@tum.de. Malteser Krankenhaus St. Franziskus-Hospital, Flensburg, Germany. angela.krackhardt@tum.de.

Tumor-derived CCL2 drives tumor growth and immunosuppression in IDH1-mutant cholangiocarcinoma

BACKGROUND AND AIMS: Isocitrate dehydrogenase 1 (IDH1)-mutant cholangiocarcinoma (CCA) is a highly lethal subtype of hepatobiliary cancer that is often resistant to immune checkpoint inhibitor therapies. We evaluated the effects of IDH1-mutations in CCA cells on the tumor immune microenvironment and identify opportunities for therapeutic intervention. APPROACH AND RESULTS: Analysis of 2,606 human CCA tumors using deconvolution of RNA-sequencing data identified decreased CD8 T cell and increased M2-like tumor-associated macrophage (TAM) infiltration in IDH1-mutant compared to IDH1-wild type tumors. To model the tumor immune microenvironment of IDH1-mutant CCA in vivo, we generated an isogenic cell line panel of mouse SB1 CCA cells containing a heterozygous IDH1 R132C (SB1mIDH1) or control (SB1WT) cells using CRISPR-mediated homology directed repair. SB1mIDH1 cells recapitulated features of human IDH1-mutant CCA including D-2-HG production and increased M2-like TAM infiltration. SB1mIDH1 cells and tumors produced increased levels of CCL2, a chemokine involved in recruitment and polarization of M2-like TAMs compared to wild type controls. In vivo neutralization of CCL2 led to decreased M2-like TAM infiltration, reduced tumor size, and improved overall survival in mice harboring SB1mIDH1 tumors. CONCLUSIONS: IDH1-mutant CCA is characterized by increased abundance of M2-like TAMs. Targeting CCL2 remodels the tumor immune microenvironment and improves outcomes in preclinical models of IDH1-mutant CCA, highlighting the role for myeloid-targeted immunotherapies in the treatment of this cancer.

Author Info: (1) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (2) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD

Author Info: (1) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (2) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (3) Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (4) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (5) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (6) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (7) Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111. (8) Tempus AI Inc., Chicago, IL 60654. (9) Tempus AI Inc., Chicago, IL 60654. (10) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (11) Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (12) Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (13) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (14) Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (15) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (16) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (17) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287. (18) Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287.

Adjuvant Immunotherapy Does Not Improve Survival in Non-small Cell Lung Cancer with Major/Complete Pathological Response after Induction Immunotherapy

OBJECTIVE: In patients with resectable non-small cell lung cancer (NSCLC), immune checkpoint inhibitor (ICI)-based regimens in both neoadjuvant and perioperative settings demonstrate survival benefit. To date, no study has compared the efficacy between pure neoadjuvant and perioperative approaches, especially in patients who achieve substantial pathological responses. METHODS: In this retrospective study, patients with clinical stage II-IIIB NSCLC who achieved either major (MPR) or complete (pCR) pathological response after induction ICI plus chemotherapy followed by resection between 2019 and 2023 were identified from multicenter databases. Inverse probability of treatment weighting-adjusted Cox regression was performed to compare disease-free survival (DFS) and overall survival (OS) between patients who did and did not receive ICIs postoperatively. RESULTS: One hundred thirty-six patients who achieved pCR and seventy-two patients who achieved MPR were enrolled. Three-quarters of them had squamous cell cancer. The inverse probability-weighted cohort represented 208 weighted patient cases (adjuvant ICI, 117; control, 91). The weighted DFS/OS rates did not differ between the adjuvant-ICI group and the control group (3-year DFS rates: 90.2% vs. 93.2%, hazard ratio [HR]= 2.47, 95% confidence interval [CI]: 0.74 to 8.22; 3-year OS rates: 89.1% vs. 93.9%, HR=2.44, 95% CI: 0.71 to 8.34). Adverse events during the postoperative ICI treatment were found in 19 of 120 patients (15.8%) and led to adjuvant ICI termination in 18 patients (15.0%). CONCLUSIONS: Adjuvant ICI does not improve survival in NSCLC patients who achieve pCR/MPR following neoadjuvant immunochemotherapy. A de-escalation strategy could be considered given the adverse events associated with postoperative ICI treatment.

Author Info: (1) Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research

Author Info: (1) Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China. (2) Key Laboratory of Carcinogenesis and Translational Research, Department of Thoracic Surgery I, Peking University Cancer Hospital and Institute, Beijing, China. (3) Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China. (4) Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China. (5) Key Laboratory of Carcinogenesis and Translational Research, Department of Thoracic Surgery I, Peking University Cancer Hospital and Institute, Beijing, China. (6) Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China. (7) Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. (8) Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China. (9) Key Laboratory of Carcinogenesis and Translational Research, Department of Thoracic Surgery I, Peking University Cancer Hospital and Institute, Beijing, China. (10) Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. Electronic address: jianglong@shchest.org.

A dendritic cell vaccine for both vaccination and neoantigen-reactive T cell preparation for cancer immunotherapy in mice

Adoptive cell transfer (ACT) using neoantigen-specific T cells is an effective immunotherapeutic strategy. However, the difficult isolation of neoantigen-specific T cells limits the clinical application of ACT. Here, we propose a method to prepare neoantigen-reactive T cells (NRT) for ACT following immunization with a tumor lysate-loaded dendritic cell (DC) vaccine. We show that the DC vaccine not only induces a neoantigen-reactive immune response in lung cancer-bearing mice in vivo, but also facilitate NRT cell preparation in vitro. Adoptive transfer of the NRTs as combinatorial therapy into DC vaccine-immunized, LL/2 tumor-bearing mice allows infiltration of the infused NRTs, as well as the enrichment of neoantigen reactive, non-ACT/NRT T cells into the tumor microenvironment with the function of these neoantigen-reactive T-cell receptors validated in vitro. In summary, we propose a method for preparing NRTs that increases ACT efficacy and paves the way to the design of personalized immunotherapies.

Author Info: (1) Department of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China. Department of Biotherapy, Ca

Author Info: (1) Department of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China. Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (2) Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (3) Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (4) Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (5) Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China. (6) Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China. (7) Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (8) Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China. (9) Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China. (10) Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (11) Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China. (12) Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. (13) Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China. Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China. (14) Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China. yl.tracy73@gmail.com. (15) Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China. dingzhenyu@scu.edu.cn.

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.