Journal Articles

Recent 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.

Serial multiomics uncovers anti-glioblastoma responses not evident by routine clinical analyses

Ling et al. provided a longitudinal multiomic view of human glioblastoma (GBM) evolution under intratumoral oncolytic viral therapy (CAN-3110), demonstrating the feasibility and importance of serial tumor sampling in studying therapeutic response. Spatial and temporal remodeling of the tumor microenvironment was mapped across 86 serial GBM biopsies from two patients. Multiomic analysis revealed therapeutic response, longitudinal and spatial reshaping of the tumor, expansion of HSV-reactive and tumor-specific T cell clonotypes, and enhanced HLA and cancer testis antigen presentation, despite indications of disease progression by MRI.

Contributed by Shishir Pant

Ling et al. provided a longitudinal multiomic view of human glioblastoma (GBM) evolution under intratumoral oncolytic viral therapy (CAN-3110), demonstrating the feasibility and importance of serial tumor sampling in studying therapeutic response. Spatial and temporal remodeling of the tumor microenvironment was mapped across 86 serial GBM biopsies from two patients. Multiomic analysis revealed therapeutic response, longitudinal and spatial reshaping of the tumor, expansion of HSV-reactive and tumor-specific T cell clonotypes, and enhanced HLA and cancer testis antigen presentation, despite indications of disease progression by MRI.

Contributed by Shishir Pant

ABSTRACT: Recurrent glioblastoma (rGBM) remains incurable. One barrier to the development of effective rGBM therapies is the difficulty in collecting posttreatment tumor tissue. Serial multiomic assays from longitudinal rGBM biopsies may uncover tumor responses to a treatment. Here, we obtained 97 serial rGBM biopsy cores over 4 months from the first two patients participating in a clinical trial of repeated intratumoral dosing of the immunotherapeutic agent CAN-3110. Multiomic analysis of the biopsy cores revealed therapeutic effects, including longitudinal and spatial reshaping of the rGBM's microenvironment, expansion of new T cell tissue-resident effector memory clonotypes against CAN-3110 epitopes and other undetermined antigens, and expression of human leukocyte antigen (HLA)-presented immunopeptides, including cancer testis antigens. Moreover, serial integrated multimodal analyses provided evidence of therapeutic responses to CAN-3110 despite traditional magnetic resonance imaging indicating progression. Clinically, the two treated patients achieved a pathologic response or stable clinical disease, respectively. These results show the value of longitudinal tissue sampling to understand rGBM's evolution during administration of an investigational therapy.

Author Info: (1) Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery and Center for Tumors of the Nervous System, Mass General Brigham Cancer Institute and Harvard Medical Sc

Author Info: (1) Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery and Center for Tumors of the Nervous System, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (2) Surgical Brain Mapping and Molecular Imaging Laboratory, Department of Neurosurgery, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (3) Department of Pathology, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. Department of Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (4) James P. Allison Institute, University of Texas MD Anderson Cancer Hospital, Houston, TX 77030, USA. (5) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (6) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (7) Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery and Center for Tumors of the Nervous System, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (8) Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery and Center for Tumors of the Nervous System, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (9) Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery and Center for Tumors of the Nervous System, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (10) Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery and Center for Tumors of the Nervous System, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (11) Surgical Brain Mapping and Molecular Imaging Laboratory, Department of Neurosurgery, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (12) Surgical Brain Mapping and Molecular Imaging Laboratory, Department of Neurosurgery, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (13) Department of Pathology, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. Department of Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (14) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (15) Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (16) Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery and Center for Tumors of the Nervous System, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. Department of Pathology, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (17) Surgical Brain Mapping and Molecular Imaging Laboratory, Department of Neurosurgery, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Broad Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3AOG4, Canada. (18) Department of Pathology, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. Department of Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (19) Department of Pathology, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. Department of Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (20) Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (21) James P. Allison Institute, University of Texas MD Anderson Cancer Hospital, Houston, TX 77030, USA. (22) James P. Allison Institute, University of Texas MD Anderson Cancer Hospital, Houston, TX 77030, USA. (23) Department of Radiation Oncology, University of Texas MD Anderson Cancer Hospital, Houston, TX 77030, USA. (24) Department of Pathology, Johns Hopkins University Medical Center, Baltimore, MD 21205, USA. (25) Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (26) Department of Neurosurgery, University of Texas MD Anderson Cancer Hospital, Houston, TX 77030, USA. (27) Department of Neuro-Oncology, University of Texas MD Anderson Cancer Hospital, Houston, TX 77030, USA. (28) Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (29) Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (30) Department of Neurology, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (31) Division of Neuro-Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Medical Center, Baltimore, MD 21205, USA. (32) Department of Neurosurgery, Johns Hopkins University Medical Center, Baltimore, MD 21205, USA. (33) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (34) Department of Pathology, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. Department of Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (35) Department of Biostatistics, University of Texas MD Anderson Cancer Hospital, Houston, TX 77030, USA. (36) Candel Therapeutics Inc., Needham, MA 02494, USA. Accelerating GBM Therapies TeamLab, Cambridge, MA 02142, USA. (37) James P. Allison Institute, University of Texas MD Anderson Cancer Hospital, Houston, TX 77030, USA. Department of Immunology, University of Texas MD Anderson Cancer Hospital, Houston, TX 77030, USA. (38) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. (39) Department of Pathology, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. Department of Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (40) Surgical Brain Mapping and Molecular Imaging Laboratory, Department of Neurosurgery, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (41) Center for Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (42) Broad Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. (43) Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery and Center for Tumors of the Nervous System, Mass General Brigham Cancer Institute and Harvard Medical School, Boston, MA 02115, USA.

Recurrent immunogenic neoantigens and their cognate T-cell receptors in treatment-resistant metastatic prostate cancer

To systematically identify recurrent clonal neoepitopes in treatment-resistant patients, Gumbert and Sagie et al. developed and applied the “Spot Neoantigens in Metastases” (SpotNeoMet) pipeline to metastatic cancer samples from the Hartwig Medical Foundation, using primary tumor samples from TCGA as control. Focusing on the common androgen receptor (AR) H875Y mutation in castration-resistant prostate cancer, they identified three neopeptides and validated their presentation and immunogenicity. Two cloned cognate TCRs were highly specific and led to killing of prostate cancer cells endogenously expressing AR H875Y in vitro and in vivo.

Contributed by Ute Burkhardt

To systematically identify recurrent clonal neoepitopes in treatment-resistant patients, Gumbert and Sagie et al. developed and applied the “Spot Neoantigens in Metastases” (SpotNeoMet) pipeline to metastatic cancer samples from the Hartwig Medical Foundation, using primary tumor samples from TCGA as control. Focusing on the common androgen receptor (AR) H875Y mutation in castration-resistant prostate cancer, they identified three neopeptides and validated their presentation and immunogenicity. Two cloned cognate TCRs were highly specific and led to killing of prostate cancer cells endogenously expressing AR H875Y in vitro and in vivo.

Contributed by Ute Burkhardt

ABSTRACT: New approaches that generate long-lasting therapeutic responses in therapy-resistant metastatic cancer patients are urgently needed. To address this challenge, we developed SpotNeoMet, a novel data-driven pipeline that systematically identifies recurrently presented neopeptides in treatment-resistant patients. We identified seven therapy resistance mutations predicted to produce neo-peptides presented by common HLAs. Using HLA-immunopeptidomics, we discovered three novel neopeptides derived from Androgen Receptor (AR) H875Y, a common metastatic castration-resistant prostate cancer (mCRPC) mutation. We validated these neoantigens as highly immunogenic and then isolated and characterized cognate T-cell receptors (TCRs) from healthy donor peripheral blood mononuclear cells. We demonstrated that AR H875Y specific TCRs are highly specific and kill prostate cancer cells presenting AR neo-peptides in vitro and in vivo. Our new pipeline identifies novel immunotherapy targets and potential treatment options for mCRPC patients. Moreover, SpotNeoMet offers a systematic route to identify 'HLA-peptide' pairs and their cognate TCRs across treatment-resistant cancers.

Author Info: (1) Weizmann Institute of Science, Rehovot, Israel. (2) Weizmann Institute of Science, Rehovot, Israel. (3) Institute for Research in Biomedicine, Barcelona, Spain. (4) Weizmann In

Author Info: (1) Weizmann Institute of Science, Rehovot, Israel. (2) Weizmann Institute of Science, Rehovot, Israel. (3) Institute for Research in Biomedicine, Barcelona, Spain. (4) Weizmann Institute of Science, Rehovot, Israel. (5) Weizmann Institute of Science, Rehovot, Israel. (6) Institute for Research in Biomedicine, Barcelona, Spain. (7) Princeton University, Princeton, NJ, United States. (8) Vall d'Hebron Institute of Oncology, Spain. (9) Weizmann Institute of Science, Rehovot, Israel. (10) Flatiron Institute, New York, NY, United States. (11) Weizmann Institute of Science, Rehovot, Israel. (12) Weizmann Institute of Science, Israel. (13) Weizmann Institute of Science, Rehovot, Israel. (14) Weizmann Institute of Science, Israel. (15) Weizmann Institute of Science, Rehovot, Israel, Israel. (16) University of Michigan-Ann Arbor, Ann Arbor, United States. (17) University of Michigan-Ann Arbor, Ann Arbor, United States. (18) Vall d'Hebron Institute of Oncology, Barcelona, Barcelona, Spain. (19) Vall d'Hebron Institute of Oncology, Spain. (20) Hadassah Medical Center, Jerusalem, Israel. (21) Weizmann Institute of Science, Israel. (22) Weizmann institute, Rehovot, Israel, Israel. (23) Weizmann Institute of Science, Rehovot, Israel, Israel. (24) Weizmann Institute of Science, Rehovot, Israel. (25) Princeton University, Princeton, NJ, United States. (26) University of British Columbia, Vancouver, British Columbia, Canada. (27) University of British Columbia, Vancouver, British Columbia, Canada. (28) University of Michigan-Ann Arbor, Ann Arbor, MI, United States. (29) University of California, San Francisco, San Francisco, CA, United States. (30) Dana-Farber Cancer Institute, Boston, MA, United States. (31) Weill Cornell Medicine, New York, NY, United States. (32) Vall d'Hebron Institute of Oncology, Barcelona, Spain. (33) Memorial Sloan Kettering Cancer Center, United States. (34) Vall d'Hebron Institute of Oncology, Spain. (35) Institute for Research in Biomedicine, Barcelona, Spain. (36) Weizmann Institute of Science, Rehovot, Israel.

Replacing cholesterol and PEGylated lipids with zwitterionic ionizable lipids in LNPs for spleen-specific mRNA translation

Zhao, Tian, Wang, et al. developed a new mRNA therapeutic cancer vaccine formulation that replaces cholesterol and PEGylated lipids in lipid nanoparticles (LNPs) with zwitterionic pyridine carboxybetaine ionizable lipids. This change increased spleen mRNA translation, reduced liver accumulation, and prevented immunogenicity against the vaccine upon repeat administration. The vaccine formulation induced specific CD8+ T cell responses, Tem and Tcm responses, and improved tumor control in murine models.

Zhao, Tian, Wang, et al. developed a new mRNA therapeutic cancer vaccine formulation that replaces cholesterol and PEGylated lipids in lipid nanoparticles (LNPs) with zwitterionic pyridine carboxybetaine ionizable lipids. This change increased spleen mRNA translation, reduced liver accumulation, and prevented immunogenicity against the vaccine upon repeat administration. The vaccine formulation induced specific CD8+ T cell responses, Tem and Tcm responses, and improved tumor control in murine models.

ABSTRACT: The spleen is emerging as a key vaccination target. However, existing lipid nanoparticles (LNPs) primarily accumulate in the liver, limiting their efficacy in vaccine therapy. The cholesterol in current LNP formulations promotes their uptake by hepatocytes, while the polyethylene glycol-modified (PEGylated) lipids induce PEG immunogenicity, further reducing the efficacy in the setting of repeated administrations. We develop a three-component (ThrCo) LNP by replacing cholesterol and PEGylated lipids in Pfizer-BioNTech LNPs with zwitterionic pyridine carboxybetaine (PyCB) ionizable lipids (ILs), achieving ~70% lower liver accumulation and a 4.5-fold increase in spleen-specific mRNA translation. PyCB ILs enhance LNP hydrophilicity, stabilizing the outer membrane to compensate for cholesterol removal. PyCB groups also exhibit strong protonation at endosomal pH, facilitating mRNA translation. The zwitterionic surface of ThrCo LNP reduces protein adsorption, thereby preventing the accelerated blood clearance effect caused by PEGylated lipids following repeated administrations. Thus, ThrCo LNP-based vaccines efficiently deliver mRNA to splenic antigen-presenting cells, boosting immune responses and improving therapeutic outcomes.

Author Info: (1) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (2) Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 1

Author Info: (1) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (2) Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA. (3) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (4) Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA. (5) Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA. (6) Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA. (7) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (8) Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA. (9) Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA. (10) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (11) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (12) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (13) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (14) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (15) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (16) Department of Biological and Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA. (17) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. (18) Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.

Combination LIGHT overexpression and checkpoint blockade disrupts the tumor immune environment impacting colorectal liver metastases

Keenan and Qiao et al. showed in mouse models of colorectal liver metastases (CRLMs, which are CTLA-4high) that anti-CTLA-4 (but not anti-PD-1) treatment controlled LIGHT-overexpressing CRLMs by generating systemic and intratumoral immune activation. scRNAseq, CyTOF, and flow cytometry showed that the LIGHT/anti-CTLA-4 combination remodeled the TME; promoted TIL migration into metastases, TLS development, activation and effector functions of T cells and DC maturation; decreased T cell exhaustion; depleted suppressive myeloid cells; and reduced Treg functionality, with some corresponding data in human CRC.

Contributed by Paula Hochman

Keenan and Qiao et al. showed in mouse models of colorectal liver metastases (CRLMs, which are CTLA-4high) that anti-CTLA-4 (but not anti-PD-1) treatment controlled LIGHT-overexpressing CRLMs by generating systemic and intratumoral immune activation. scRNAseq, CyTOF, and flow cytometry showed that the LIGHT/anti-CTLA-4 combination remodeled the TME; promoted TIL migration into metastases, TLS development, activation and effector functions of T cells and DC maturation; decreased T cell exhaustion; depleted suppressive myeloid cells; and reduced Treg functionality, with some corresponding data in human CRC.

Contributed by Paula Hochman

ABSTRACT: Colorectal cancer and liver metastases are a leading cause of cancer-related mortality. Overexpression of the immunostimulatory cytokine TNFSF14/LIGHT associates with improved survival and correlates with increased tumor-infiltrating lymphocytes in patients and a clinically relevant model of colorectal liver metastases. We demonstrate that LIGHT monotherapy activates T cells, but also induces T cell exhaustion and the recruitment of immunosuppressive elements. As colorectal liver metastases exhibit high levels of CTLA-4 expression, we combined LIGHT overexpression with anti-CTLA-4, leading to complete tumor control. The combination functions by homing tumor-infiltrating lymphocytes, inducing tumor antigen-specific T cells, and reversing T cell exhaustion. Whereas both LIGHT overexpression and anti-CTLA-4 increase tumor-promoting macrophages, the combination eliminates this population. The ability of LIGHT overexpression combined with CTLA-4 inhibition to reverse T cell exhaustion and myeloid cell suppression is supported by analysis of complementary patient cohorts and has strong clinical relevance, especially given that liver metastases contribute to immunotherapy resistance across various cancer types.

Author Info: (1) Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA 94143, USA. (2) Department of Surgery, Division of Surgical

Author Info: (1) Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA 94143, USA. (2) Department of Surgery, Division of Surgical Oncology, University of California, San Francisco, San Francisco, CA 94143, USA. (3) Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA. (4) Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA. (5) Departments of Otolaryngology-Head and Neck Surgery and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA. (6) Department of Surgery, Division of Surgical Oncology, University of California, San Francisco, San Francisco, CA 94143, USA. (7) Department of Microbiology & Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA. (8) Department of Surgery, Division of Surgical Oncology, University of California, San Francisco, San Francisco, CA 94143, USA. (9) Department of Surgery, Division of Surgical Oncology, University of California, San Francisco, San Francisco, CA 94143, USA. (10) Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA. (11) Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA 94143, USA. Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA. (12) Departments of Otolaryngology-Head and Neck Surgery and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA. Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA. Chan Zuckerberg Biohub, San Francisco, CA 94158, USA. (13) Department of Microbiology & Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA. (14) Department of Surgery, Division of Surgical Oncology, University of California, San Francisco, San Francisco, CA 94143, USA.

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.