Journal Articles

Biselective remodeling of the melanoma tumor microenvironment prevents metastasis and enhances immune activation in mouse models

The extracellular matrix (ECM) plays a crucial role in supporting metastasis in solid malignancies, yet effective ECM-targeted therapies remain scarce. Here, we introduce a dual-targeting strategy to combat melanoma by leveraging bispecific agents that disrupt key ECM and tumor-associated pathways. Building on the inhibitory properties of lysyl oxidase-propeptide (LOX-PP), we engineered biselective decoys that simultaneously target the collagen cross-linking enzyme LOX and heat shock protein 70 (HSP70), both of which are up-regulated during melanoma progression in both human and mouse models. This dual-targeting strategy offers a new avenue for disrupting ECM-driven tumor progression and enhancing therapeutic efficacy. Administered to mouse models of melanoma, the decoys reduced tumor burden and circulating melanoma cells by inhibiting proliferation and lung metastasis. Mechanistically, the decoys suppressed cancer-supporting ECM organization, inhibited ECM-remodeling pathways and associated enzymes, and reshaped the tumor immune microenvironment. The treatment modulated immune responses by enhancing neutrophil, B cell, and CD8(+) T cell infiltration. In combination with immune check point inhibitor, the decoys further promoted melanoma killing by CD8(+) T cells. The decoys efficiently bound multiple human tumors expressing LOX(+)/HSP70(+) ex vivo. These findings highlight the potential of dual inhibition as a potential strategy for remodeling melanoma and other tumor microenvironments and enhancing immunotherapy efficacy.

Author Info: (1) Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel. Department of Agricultural Development, Agrofood an

Author Info: (1) Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel. Department of Agricultural Development, Agrofood and Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, Psachna 34400, Greece. (2) Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel. (3) Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel. (4) Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel. (5) Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel. (6) Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel. (7) Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel. (8) Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel. (9) Institute of Pathology, E. Wolfson Medical Center, Holon 58100, Israel. (10) Institute of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel. (11) Institute of Pathology, E. Wolfson Medical Center, Holon 58100, Israel. (12) Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel. Department of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel. (13) Institute of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel. Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel. (14) Univ. Lyon, Institut de Chimie et Biochimie MolŽculaires et SupramolŽculaires (ICBMS), UMR 5246, University Lyon 1, CNRS, Villeurbanne F-69622, France. (15) Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel. (16) Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel.

Dendritic cell progenitors engineered to express extracellular-vesicle-internalizing receptors enhance cancer immunotherapy in mouse models

Cancer immunotherapy using dendritic cells (DC) pulsed ex vivo with tumour antigens is considered safe, but its clinical efficacy is generally modest. Here we engineer DC progenitors (DCP), which can replenish conventional type 1 DCs (cDC1) in mice, to constitutively express IL-12 together with a non-signalling chimeric receptor, termed extracellular vesicle-internalizing receptor (EVIR). By binding to a bait molecule (GD2 disialoganglioside) expressed on cancer cells and their EVs, the EVIR enforces EV internalization by cDC1 to promote their cross-dressing with preformed, tumour-derived MHCI-peptide complexes. Upon systemic deployment to mice, the engineered DCPs cause only mild and transient elevation of liver enzymes, acquire tumour-derived material, engage tumour-specific T cells, and enhance the efficacy of PD-1 blockade in an immunotherapy-resistant melanoma model comprising both GD2-positive and -negative cancer cells, without the need for ex vivo antigen pulsing. These results indicate that EVIR-engineered DCPs may avert the positive selection of antigen-negative cancer cells, potentially addressing a critical limitation of immunotherapies targeting defined tumour antigens.

Author Info: (1) Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland. Agora Cancer

Author Info: (1) Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland. Agora Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center LŽman (SCCL), Lausanne, Switzerland. (2) Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland. Agora Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center LŽman (SCCL), Lausanne, Switzerland. (3) Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland. Agora Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center LŽman (SCCL), Lausanne, Switzerland. (4) Laboratory of Metabolic Signaling, Institute of Bioengineering, EPFL, Lausanne, Switzerland. (5) Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland. Agora Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center LŽman (SCCL), Lausanne, Switzerland. (6) Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland. Agora Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center LŽman (SCCL), Lausanne, Switzerland. (7) Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland. Agora Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center LŽman (SCCL), Lausanne, Switzerland. (8) Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland. Agora Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center LŽman (SCCL), Lausanne, Switzerland. (9) Animal Modeling Facility, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands. (10) Animal Modeling Facility, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands. (11) Department of Oncology, University of Lausanne (UNIL), Lausanne, Switzerland. Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland. (12) Agora Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center LŽman (SCCL), Lausanne, Switzerland. Department of Oncology, Geneva University Hospital (HUG), Geneva, Switzerland. Center for Translational Research in Onco-Hematology, University of Geneva (UNIGE), Geneva, Switzerland. (13) Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland. michele.depalma@epfl.ch. Agora Cancer Research Center, Lausanne, Switzerland. michele.depalma@epfl.ch. Swiss Cancer Center LŽman (SCCL), Lausanne, Switzerland. michele.depalma@epfl.ch.

A next-generation anti-CTLA-4 probody mitigates toxicity and enhances anti-tumor immunity in mice

CTLA-4 is a promising target for immune checkpoint inhibition in cancer therapy, with CTLA-4 blockade achieving prolonged overall survival for responding patients. However, the progressively elevated doses of anti-CTLA-4 agents, aimed at achieving better efficacy, result in increased toxicities, limiting their clinical applications. Here, we generate a prodrug design of the anti-CTLA-4 antibody, named ProCTLA-4, by folding the Fab fragment of the antibody in a tumor-associated protease-based manner. In preclinical mouse models, ProCTLA-4 effectively depletes suppressive regulatory T cells within the tumor microenvironment and enhances tumor-associated antigen-specific CD8(+) T cell responses, while exhibiting reduced toxicity compared to currently available CTLA-4 blockade approaches. Furthermore, compared to the currently used Probody therapeutics for anti-CTLA-4 (BMS986288), ProCTLA-4 has more advantages in efficacy amplification, such as in poor immunogenic melanoma. Our design establishes an alternative paradigm for antibody agents that limits the emergence of immune-related adverse events (irAE) while increasing therapeutic efficacy.

Author Info: (1) School of Basic Medical Sciences, Tsinghua University, Beijing, China. State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijin

Author Info: (1) School of Basic Medical Sciences, Tsinghua University, Beijing, China. State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China. (2) School of Basic Medical Sciences, Tsinghua University, Beijing, China. State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China. (3) School of Basic Medical Sciences, Tsinghua University, Beijing, China. State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China. (4) School of Basic Medical Sciences, Tsinghua University, Beijing, China. State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China. (5) School of Basic Medical Sciences, Tsinghua University, Beijing, China. State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China. (6) School of Basic Medical Sciences, Tsinghua University, Beijing, China. State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China. (7) School of Basic Medical Sciences, Tsinghua University, Beijing, China. yangxinfu@tsinghua.edu.cn. State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China. yangxinfu@tsinghua.edu.cn. Changping Laboratory, Changping District, Beijing, China. yangxinfu@tsinghua.edu.cn. (8) School of Basic Medical Sciences, Tsinghua University, Beijing, China. wywang2022@tsinghua.edu.cn. State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China. wywang2022@tsinghua.edu.cn.

IL-2/IL-15 signaling induces NK cell production of FLT3LG augmenting anti-PD-1 immunotherapy

Natural killer (NK) cells play a critical role in anti-cancer immunity through their direct cytotoxicity and production of cytokines, such as Flt3L. NK cell production of Flt3L controls conventional type I dendritic cell (cDC1) abundance in the tumor and promotes protective immune responses. Here, we show that NK cell production of Flt3l in the tumor is regulated by activation, and that activation by IL-2 and IL-15 uniquely induced Flt3L expression in NK cells. In melanoma, IL-2 signaling in NK cells led to increased Flt3L production, which boosted cDC1 abundance in the tumor and improved anti-PD-1 immunotherapy response. Further, NK cell subsets differentially regulated Flt3L in the tumor, with CD11b-CD27+ NK cells in mouse tumors enriched for IL-2-family signaling and upregulated Flt3l upon activation. Consistently, human CD56brightCD16- NK cells more strongly correlated with cDC1 and FLT3LG expression than other NK cell subsets across multiple human melanoma datasets and cancer indications. This mechanistic study of NK cell regulation of FLT3LG and control of the NK cell-cDC1 axis provides insights and strategies for the development of more effective cancer immunotherapies.

Author Info: (1) Fred Hutchinson Cancer Center, United States. (2) Fred Hutchinson Cancer Center, United States. (3) Fred Hutchinson Cancer Center, Seattle, WA, United States. (4) Massachusetts

Author Info: (1) Fred Hutchinson Cancer Center, United States. (2) Fred Hutchinson Cancer Center, United States. (3) Fred Hutchinson Cancer Center, Seattle, WA, United States. (4) Massachusetts General Hospital Cancer Center, Boston, Massachusetts, United States. (5) Fred Hutchinson Cancer Center, Seattle, WA, United States. (6) Fred Hutchinson Cancer Center, Seattle, United States. (7) Fred Hutchinson Cancer Center, United States. (8) Fred Hutchinson Cancer Center, Seattle, WA, United States. (9) Fred Hutchinson Cancer Center, Seattle, France. (10) Fred Hutchinson Cancer Center, Seattle, WA, United States.

Aberrant expression of SLAMF6 constitutes a targetable immune escape mechanism in acute myeloid leukemia

Immunotherapy has shown limited success in acute myeloid leukemia (AML), indicating an incomplete understanding of the underlying immunoregulatory mechanisms. Here we identify an immune evasion mechanism present in 60% of AML cases, wherein primitive AML cells aberrantly express the lymphoid surface protein SLAMF6 (signaling lymphocyte activation molecule family member 6). Knockout of SLAMF6 in AML cells enables T cell activation and highly efficient killing of leukemia cells in coculture systems, demonstrating that SLAMF6 protects AML cells from recognition and elimination by the immune system in a mode analogous to the programmed cell death protein-ligand (PDL1/PD1) axis. Targeting SLAMF6 with an antibody against the SLAMF6 dimerization site inhibits the SLAMF6-SLAMF6 interaction and induces T cell activation and killing of AML cells both in vitro and in humanized in vivo models. In conclusion, we show that aberrant expression of SLAMF6 is a common and targetable immune escape mechanism that could pave the way for immunotherapy in AML.

Author Info: (1) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. carl.sanden@med.lu.se. (2) Division of Clinical Genetics, Department of Laborat

Author Info: (1) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. carl.sanden@med.lu.se. (2) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. Department of Hematology, Oncology and Radiation Physics, SkŒne University Hospital, Lund, Sweden. (3) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. (4) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. (5) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. (6) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. (7) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. (8) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. (9) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. (10) Department of Hematology, Oncology and Radiation Physics, SkŒne University Hospital, Lund, Sweden. (11) Department of Hematology, Oncology and Radiation Physics, SkŒne University Hospital, Lund, Sweden. (12) Department of Immunotechnology, Lund University, Lund, Sweden. SciLifeLab Drug Discovery and Development Platform, Lund University, Lund, Sweden. (13) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. (14) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. Department of Clinical Genetics, Pathology, and Molecular Diagnostics, SkŒne University Hospital, Region SkŒne, Lund, Sweden. (15) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. (16) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. Department of Clinical Genetics, Pathology, and Molecular Diagnostics, SkŒne University Hospital, Region SkŒne, Lund, Sweden. (17) Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden. thoas.fioretos@med.lu.se. Department of Clinical Genetics, Pathology, and Molecular Diagnostics, SkŒne University Hospital, Region SkŒne, Lund, Sweden. thoas.fioretos@med.lu.se.

Local delivery of IL-15 and anti-PD-L1 nanobody by in vitro-transcribed circILNb elicits superior antitumor immunity in cold tumors

The clinical translation of combined immunocytokine (IC) and immune checkpoint inhibitor (ICI) is constrained by relapse of advanced malignancies, systemic toxicities, and prohibitive research and synthesis costs. In this study, the circCV-B3 vector is constructed to enable scarless circular RNA (circRNA) engineering. The circILNb, engineered via the circCV-B3 vector, enables co-encoding of interleukin-15 (IL-15) and anti-PD-L1 nanobody (Nb). The circILNb is purified by biotin-avidin purification system (BAPS) and is encapsulated within lipid nanoparticles (LNPs). Intratumoral circILNb administration achieves in situ protein expression, achieving local tumor control. Furthermore, dendritic cells (DCs) load circILNb and migrate to tumor-draining lymph node (tdLN), where they prime antigen-specific CD8(+) T cell activation, eliciting a robust systemic immune response. These findings highlight the potential of circCV-B3 vector and BAPS as a methodology for circRNA engineering and substantiate circILNb as non-protein-based therapeutic strategy for tumor immunotherapy.

Author Info: (1) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (2) Department of Clinical Biochemistry, Fac

Author Info: (1) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (2) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (3) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (4) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (5) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (6) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (7) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (8) College of Education and Science, Chongqing Normal University, Chongqing 400047, China. (9) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (10) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (11) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. (12) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. Electronic address: yangmingzhen0807@126.com. (13) National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. Electronic address: qmzou2007@163.com. (14) Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing 400038, China. Electronic address: lianjiqin@tmmu.edu.cn.

CRATER tumor niches facilitate CD8+ T cell engagement and correspond with immunotherapy success

ABSTRACT: T cell-mediated tumor killing underlies immunotherapy success. Here, we used long-term in vivo imaging and high-resolution spatial transcriptomics of zebrafish endogenous melanoma, as well as multiplex imaging of human melanoma, to identify domains facilitating the immune response during immunotherapy. We identified cancer regions of antigen presentation and T cell engagement and retention (CRATERs) as pockets at the stroma-melanocyte boundaries of zebrafish and human melanoma. CRATERs are rich in antigen-recognition molecules, harboring the highest density of CD8(+) T cells in tumors. In zebrafish, CD8(+) T cells formed prolonged interactions with melanoma cells within CRATERs, characteristic of antigen recognition. Following immunostimulatory treatment, CRATERs expanded, becoming the major sites of activated CD8(+) T cell accumulation and tumor killing. In humans, elevation in CRATER density in biopsies following immune checkpoint blockade (ICB) therapy correlated with a clinical response to therapy. CRATERs are structures that show active tumor killing and may be useful as a diagnostic indicator for immunotherapy success.

Author Info: (1) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Ho

Author Info: (1) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA. (2) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA. (3) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA. (4) Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center at Harvard, Boston, MA 02115, USA; Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA. (5) Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (6) Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (7) Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (8) Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (9) Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (10) Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (11) Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (12) Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA. (13) Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA. (14) Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA. (15) Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (16) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA. (17) Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA. (18) Department of Physics, University of Chicago, Chicago, IL 60637, USA. (19) Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (20) Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (21) Harvard Center for Biological Imaging, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA. (22) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA. (23) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA. (24) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA. (25) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA. (26) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA. (27) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA. (28) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA. (29) Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA. (30) Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA. (31) Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA. (32) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA. (33) Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA. (34) Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA. (35) Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA. (36) Department of Bioengineering and Applied Sciences, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA. (37) Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (38) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA. (39) Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. (40) Ludwig Center at Harvard, Boston, MA 02115, USA; Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA. (41) Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Parker Institute for Cancer Immunotherapy, Boston, MA, USA. (42) Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA. (43) Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA. (44) Harvard Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA 02138, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA. Electronic address: leonard.zon@enders.tch.harvard.edu.

Primate resident memory T cells activate humoral and stromal immunity

Joag et al. demonstrated that systemic T cell-based vaccines generate robust T cell surveillance throughout the body, integrating cellular and humoral immunity. Intravenous heterologous prime-boost vaccination in macaques established SIV-gag–specific CD8⁺ memory (including resident memory [Trm]), T cells across more than 30 lymphoid, mucosal, and visceral tissues. Local Trm reactivation in the reproductive tract mucosa activated stromal, endothelial, and innate lymphoid cells, amplifying interferon-driven antiviral programs. These signals recruited CD4⁺ T cells with host-defense signatures, and mobilized B and plasma cells.

Contributed by Shishir Pant

Joag et al. demonstrated that systemic T cell-based vaccines generate robust T cell surveillance throughout the body, integrating cellular and humoral immunity. Intravenous heterologous prime-boost vaccination in macaques established SIV-gag–specific CD8⁺ memory (including resident memory [Trm]), T cells across more than 30 lymphoid, mucosal, and visceral tissues. Local Trm reactivation in the reproductive tract mucosa activated stromal, endothelial, and innate lymphoid cells, amplifying interferon-driven antiviral programs. These signals recruited CD4⁺ T cells with host-defense signatures, and mobilized B and plasma cells.

Contributed by Shishir Pant

ABSTRACT: CD8(+) resident memory T (Trm) cells comprise a small population of frontline sentinels compared with the large tissues they surveil, making outsized contributions to immune protection from infection. Here, we interrogated mechanisms of Trm cell function in primates. Intravenous immunization of macaques with a simian immunodeficiency virus (SIV)-gag-containing heterologous prime-boost-boost vaccine established memory T cells in >30 tissues, including visceral and mucosal compartments. Upon in vivo reactivation in the reproductive tract, antigen-sensing CD8(+) Trm activated local stromal, parenchymal, and innate and adaptive immune cells. Stromal and parenchymal cells accentuated leukocyte migration and antiviral defenses. B and plasma cells mobilized into the vaginal mucosa, and bloodborne CD4(+) T cells were recruited and adopted a host-defense program. Our findings demonstrate that systemic vaccination promotes a Trm cell response in barrier compartments and that Trm cells repurpose abundant neighboring stromal, parenchymal, and immune cells to amplify alarm signals and activate diverse host defenses.

Author Info: (1) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. Electronic address: vineet.joag@seattlechildrens.org. (2)

Author Info: (1) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. Electronic address: vineet.joag@seattlechildrens.org. (2) Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA. (3) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (4) Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30322, USA. (5) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (6) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (7) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (8) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (9) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (10) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (11) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (12) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (13) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (14) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (15) Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30322, USA. (16) Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30322, USA. (17) Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, USA. (18) Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6G 2V4, Canada. (19) Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA. (20) Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA. (21) Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota, Minneapolis, MN, USA. (22) Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA. (23) Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA. (24) Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, USA. (25) Center for Innate Immunity and Inflammation, Pelotonia Institute for Immuno-oncology, the James Comprehensive Cancer Center, Department of Surgery, Ohio State University, Columbus, OH, USA. (26) Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA. (27) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. (28) Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota, Minneapolis, MN, USA. (29) Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA. (30) Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30322, USA. (31) Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA. Electronic address: masopust@umn.edu.

Anticancer immune responses are hindered by cis interaction of inhibitory checkpoint SIRPα

Tang et al. showed that phagocytosis suppression by Signal Regulatory Protein α (SIRPα) depends not only on trans CD47 binding and phosphatase signaling, but also on cis interactions with CD18 (β2 integrin) on the macrophage surface. Distinct SIRPα residues interacted with CD18 and CD47. SIRPα–CD18 interaction prevented Mac-1 (αMβ2, CD18/CD11b) activation, which is required for phagocytosis in synergy with the SIRPα–CD47 interaction. In mouse tumor models, treatment with anti-SIRPα bispecific Abs blocking both CD47 and CD18 interactions caused a greater increase in phagocytosis and antitumor activity than monospecific anti-SIRPα Abs.

Contributed by Paula Hochman

Tang et al. showed that phagocytosis suppression by Signal Regulatory Protein α (SIRPα) depends not only on trans CD47 binding and phosphatase signaling, but also on cis interactions with CD18 (β2 integrin) on the macrophage surface. Distinct SIRPα residues interacted with CD18 and CD47. SIRPα–CD18 interaction prevented Mac-1 (αMβ2, CD18/CD11b) activation, which is required for phagocytosis in synergy with the SIRPα–CD47 interaction. In mouse tumor models, treatment with anti-SIRPα bispecific Abs blocking both CD47 and CD18 interactions caused a greater increase in phagocytosis and antitumor activity than monospecific anti-SIRPα Abs.

Contributed by Paula Hochman

ABSTRACT: Signal regulatory protein α (SIRPα) is a macrophage inhibitory receptor that limits phagocytosis and antitumor activity by interacting in trans with CD47 on tumor cells. Here, we found that a component of SIRPα's inhibitory function occurred independently of CD47. Inhibition occurred because of interactions between SIRPα and CD18 (β2 integrin) in cis on the surface of macrophages, involving SIRPα amino acids distinct from those implicated in the SIRPα-CD47 interaction. This cis interaction prevented activation of CD18, which is necessary for phagocytosis. The combined blockade of SIRPα-CD18 and SIRPα-CD47 was essential for maximizing phagocytosis and suppression of tumor growth in vivo. Thus, inhibitory immune checkpoints such as SIRPα suppress cell activation through a mechanism targeting CD18 in cis, which occurs in addition to engagement by their inhibitory checkpoint ligands in trans. This dual mode of action should be considered when developing inhibitory checkpoint blockades for immunotherapy.

Author Info: (1) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. Cancer Center, Faculty of Health Sciences, University of Macau, Macau S

Author Info: (1) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China. MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China. (2) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. (3) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. (4) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. Department of Medicine, McGill University, MontrŽal, Canada. (5) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. Department of Medicine, McGill University, MontrŽal, Canada. (6) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. Department of Medicine, McGill University, MontrŽal, Canada. (7) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. (8) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. (9) Laboratory of Molecular Oncology, Institut de recherches cliniques de MontrŽal (IRCM), MontrŽal, Canada. Department of Medicine, McGill University, MontrŽal, Canada. Department of Medicine, University of MontrŽal, MontrŽal, Canada.

Collagen-disrupting attIL12 TIL therapy boosts deep T cell infiltration via dual signaling activation and CCKAR reduction in sarcomas

Across clinical sarcomas, collagen expression increased with tumor stage and associated with reduced patient survival. To treat ECM-rich tumors, Hu and Singh et al. modified TILs to express a cell-surface vimentin (CSV, sarcoma antigen)-targeting peptide linked to IL-12 (attIL12-TILs). Compared to standard TILs, attIL12-TILs improved IFNγ production, reduced tumor collagen expression, and boosted infiltration and tumor control in mouse sarcoma models. Inhibiting signaling by either TCRs or CSV abrogated these effects. Mechanistically, attIL12-TILs reduced tumor cell collagen levels via IFNγ inhibition of TGFβ-SMAD3 and CCKAR-pAKT pathways.

Contributed by Alex Najibi

Across clinical sarcomas, collagen expression increased with tumor stage and associated with reduced patient survival. To treat ECM-rich tumors, Hu and Singh et al. modified TILs to express a cell-surface vimentin (CSV, sarcoma antigen)-targeting peptide linked to IL-12 (attIL12-TILs). Compared to standard TILs, attIL12-TILs improved IFNγ production, reduced tumor collagen expression, and boosted infiltration and tumor control in mouse sarcoma models. Inhibiting signaling by either TCRs or CSV abrogated these effects. Mechanistically, attIL12-TILs reduced tumor cell collagen levels via IFNγ inhibition of TGFβ-SMAD3 and CCKAR-pAKT pathways.

Contributed by Alex Najibi

ABSTRACT: Tumor-targeted T cell therapies of various types have been booming, but T cell therapy is limited by its inability to penetrate the collagen barrier surrounding tumors. The destruction of tumor collagen is significant because collagen both suppresses T cells and contributes to the formation of the extracellular matrix. Our previously reported cell-surface vimentin (CSV)-targeted and membrane-anchored interleukin 12-armed (attIL12) T cells can reduce collagen production by killing cancer-associated fibroblasts, but fail to reduce collagen expression by tumor cells, resulting in resistance to attIL12-T cell treatment. In this study, we found that CCKAR directly boosts collagen production by tumor cells in vitro and in vivo. attIL12-modified tumor-infiltrating lymphocytes (TILs) disabled collagen production by CCKAR-high autologous tumor cells in vitro and sarcoma patient-derived xenografts (PDXs) in vivo. This disruption of collagen production by tumor cells by attIL12-TILs overcomes resistance to attIL12-T cell treatment and required a simultaneous interaction between the CSV on autologous tumor cells, which is targeted by attIL12, and human leukocyte antigen-T cell receptor on attIL12-TILs; When either interaction was abrogated, collagen production and CCKAR expression were not shut down. Mechanistically, the interaction between attIL12-TILs and autologous tumor cells induced interferon gamma production synergistically, which in combination with CCKAR downregulation reduced collagen expression through suppression of both transforminggrowth factor beta-stimulated SMAD activation and CCKAR-AKT signaling. Diminishing collagen expression from tumor cells significantly increased T cell infiltration and improved tumor growth inhibition in PDX sarcomas. Thus, this attIL12-TIL therapy holds great clinical potential for boosting T cell infiltration in high-grade, collagen-rich tumors.

Author Info: (1) Division of Pediatrics, Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030. (2) Division of Pediatrics, Department of Pedia

Author Info: (1) Division of Pediatrics, Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030. (2) Division of Pediatrics, Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030. (3) Division of Pediatrics, Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030. (4) Division of Pediatrics, Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030. (5) Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030. (6) Division of Pediatrics, Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030. (7) Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030. (8) Division of Pediatrics, Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030. (9) Division of Pediatrics, Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.

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.