Journal Articles

IL-9 as a naturally orthogonal cytokine with optimal JAK/STAT signaling for engineered T cell therapy

Taking advantage of low IL-9R expression in normal tissues, Jiang et al. engineered T cells with wild-type IL-9R. After transfer into tumor-bearing mice in combination with IL-9, which was well tolerated, IL-9R-engineered T cells showed enhanced infiltration, stemness, and antitumor activity compared to similar studies with orthogonal IL-9R. The IL-9–IL-9R axis activated STAT1/3/5 and (unexpectedly) recruited STAT4, and was highly sensitive to alterations in IL-9R signal strength or pSTAT stoichiometry. Levels of pSTAT1 determined differentiation towards stem/memory or terminal effector states. Similar results were observed CAR T cells.

Contributed by Lauren Hitchings

Taking advantage of low IL-9R expression in normal tissues, Jiang et al. engineered T cells with wild-type IL-9R. After transfer into tumor-bearing mice in combination with IL-9, which was well tolerated, IL-9R-engineered T cells showed enhanced infiltration, stemness, and antitumor activity compared to similar studies with orthogonal IL-9R. The IL-9–IL-9R axis activated STAT1/3/5 and (unexpectedly) recruited STAT4, and was highly sensitive to alterations in IL-9R signal strength or pSTAT stoichiometry. Levels of pSTAT1 determined differentiation towards stem/memory or terminal effector states. Similar results were observed CAR T cells.

Contributed by Lauren Hitchings

ABSTRACT: Cytokines and their receptors enable precise tuning of T cell function. Leveraging this biology holds tremendous promise for optimizing antitumor immunity. Arming T cells with a synthetically orthogonal interleukin (IL)-9 receptor (o9R), for instance, permits facile engraftment and potent anti-tumor functions. Exploiting the paucity of wild-type IL-9R expression and the safety of high doses of IL-9, here, we showed that, compared with o9R, T cells engineered with wild-type IL-9R exhibited superior tissue infiltration, stemness, and anti-tumor activity. These qualities were consistent with a stronger Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signal, which included canonically IL-12-driven STAT4 in addition to STAT1/3/5. IL-9R T cells were exquisitely sensitive to perturbations of proximal signaling, including structure-guided attenuation, amplification, and rebalancing of JAK/STAT signals. Biased IL-9R mutants showed that STAT1 acts as a rheostat between stem-like and effector states. In summary, we identify IL-9/IL-9R as a naturally orthogonal cytokine-receptor pair with an optimal JAK/STAT signaling profile for engineered T cell therapy.

Author Info: (1) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Molecular and Cellular Physiology and Structural Biology, Stan

Author Info: (1) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. (2) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA. (3) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94143, USA; Stanford Center for Cancer Cell Therapy, Stanford University School of Medicine, Stanford, CA 94143, USA. (4) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA. (5) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA. (6) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA. (7) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA. (8) Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. (9) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA. (10) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA. (11) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA. (12) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA. (13) Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. (14) Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. (15) Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. (16) Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA 90095, USA. (17) Department of Pathology & Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Pediatric Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA 90095, USA. (18) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94143, USA. (19) Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, Suite D3500, San Francisco, CA 94129, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA. (20) Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Center for Cancer Cell Therapy, Stanford University School of Medicine, Stanford, CA 94143, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94143, USA. Electronic address: akalbasi@stanford.edu.

Macrophage-targeted immunocytokine leverages myeloid, T, and NK cell synergy for cancer immunotherapy

Setting out to reprogram TAMs and stimulate adjacent T cells in the TME, von Locquenghien, Zwicky, Cie, et al. developed a dual-targeting MiTE strategy. The developed MiTE consisted of a TAM-targeting anti-TREM2 antibody with an attached IL-2 variant with high affinity for IL-2Rβ. The IL-2 variant had a blocking domain that rendered it inactive, but could be activated in the TME through TAM-produced proteases. MiTE treatment remodeled the immune TME in mouse models, with shifts in TAMs to inflammatory monocytic states, and activation of T cells. Therapy resulted in reduced tumor growth, and effects were more pronounced when therapy was combined with ICB. Similar effects were observed ex vivo in patient-derived tumor fragments.

Setting out to reprogram TAMs and stimulate adjacent T cells in the TME, von Locquenghien, Zwicky, Cie, et al. developed a dual-targeting MiTE strategy. The developed MiTE consisted of a TAM-targeting anti-TREM2 antibody with an attached IL-2 variant with high affinity for IL-2Rβ. The IL-2 variant had a blocking domain that rendered it inactive, but could be activated in the TME through TAM-produced proteases. MiTE treatment remodeled the immune TME in mouse models, with shifts in TAMs to inflammatory monocytic states, and activation of T cells. Therapy resulted in reduced tumor growth, and effects were more pronounced when therapy was combined with ICB. Similar effects were observed ex vivo in patient-derived tumor fragments.

ABSTRACT: Tumor-associated macrophages (TAMs) expressing the myeloid checkpoint TREM2 are key immunosuppressive cells in the tumor microenvironment (TME), driving tumor progression and contributing to poor prognosis in cancer patients. Due to their pivotal role, TAMs have emerged as a promising target for immunotherapies. However, current TAM-targeting monotherapies show limited efficacy, highlighting the need for strategies engaging multiple immune modalities. Here, we developed myeloid-targeted immunocytokines and natural killer (NK)/T cell enhancers (MiTEs) harnessing myeloid and lymphoid synergy for immunotherapy. MiTEs are trans-acting immunocytokines with tumor-specific activation, allowing dual targeting of TAMs and lymphocytes by TREM2 antagonism and cytotoxic effector cell activation through interleukin (IL)-2. To avoid off-target toxicities, MiTEs contain an IL-2 masking moiety, which is cleaved by a TAM-specific protease. MiTEs demonstrate high efficacy in preclinical tumor models through extensive immune reprogramming spanning TAM, T, and NK cell compartments. MiTEs show transformative potential for treating solid cancers by inducing potent multi-axis anti-tumor immunity while minimizing toxicities.

Author Info: (1) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (2) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001,

Author Info: (1) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (2) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (3) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (4) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (5) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (6) Immunai, New York, NY 10016, USA. (7) Immunai, New York, NY 10016, USA. (8) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel; Rheumatology Department, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel. (9) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (10) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (11) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (12) Immunai, New York, NY 10016, USA. (13) Immunai, New York, NY 10016, USA. (14) Immunai, New York, NY 10016, USA. (15) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. (16) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel; Immunai, Ramat Gan 5252213, Israel. (17) Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. Electronic address: ido.amit@weizmann.ac.il.

Predominant mutated non-canonical tumor-specific antigens identified by proteogenomics demonstrate immunogenicity and tumor suppression in CRC

Xiang et al. integrated whole-genome, transcriptomics, and MHC-I immunopeptidomics analyses to identify tumor-specific antigens from non-coding regions in colorectal cancer samples. Across 10 paired samples, over 80% of 96 MHC-I-presented neo-epitopes originated from intergenic and intronic regions. Hypermutated tumors showed the highest burden of non-canonical neo-epitopes, while non-hypermutated tumors relied on coding alterations and alternative splicing. In the subcutaneous MC38 model, multi-epitope vaccines containing mutated non-canonical neo-epitopes effectively activated CD8+ T cells and suppressed tumor growth in a CD8+ T cell-dependent manner.

Contributed by Shishir Pant

Xiang et al. integrated whole-genome, transcriptomics, and MHC-I immunopeptidomics analyses to identify tumor-specific antigens from non-coding regions in colorectal cancer samples. Across 10 paired samples, over 80% of 96 MHC-I-presented neo-epitopes originated from intergenic and intronic regions. Hypermutated tumors showed the highest burden of non-canonical neo-epitopes, while non-hypermutated tumors relied on coding alterations and alternative splicing. In the subcutaneous MC38 model, multi-epitope vaccines containing mutated non-canonical neo-epitopes effectively activated CD8+ T cells and suppressed tumor growth in a CD8+ T cell-dependent manner.

Contributed by Shishir Pant

ABSTRACT: Tumor-specific antigens (TSAs) are crucial for activating T cells against cancer, but traditional discovery methods focusing on exonic mutations overlook non-canonical TSAs from non-coding regions. We employed an integrative proteogenomic strategy combining whole-genome and RNA sequencing with immunoprecipitation mass spectrometry to comprehensively explore TSA generation in colorectal cancer patients. Analysis of 10 paired tumor samples identified 96 mutated major histocompatibility complex class I-presented neo-epitopes, with 80.21% originating from non-coding regions. In hypermutated tumors with high mutational burden, neo-epitopes predominantly arose from intergenic and intronic areas, while in non-hypermutated tumors with low mutational burden, they mainly stemmed from coding variations and alternative splicing events. Functional validation in mouse models demonstrated that mutated non-canonical neo-epitopes effectively activated CD8(+) T cells and significantly suppressed tumor growth. These findings underscore the importance of considering the entire genomic landscape in TSA discovery, suggesting new avenues for personalized immunotherapy.

Author Info: (1) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Hangzhou 310030,

Author Info: (1) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China. (2) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China. (3) National Center for Nanoscience and Technology, Beijing 100190, China. (4) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen 518083, China. (5) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China. (6) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Hangzhou 310030, China. (7) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Hangzhou 310030, China. (8) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China. (9) BGI Research, Shenzhen 518083, China. (10) BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China. (11) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen 518083, China. (12) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen 518083, China. (13) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen 518083, China. (14) The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510655, China. (15) BGI Research, Shenzhen 518083, China; BGI Research, Changzhou 213299, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGl-Shenzhen, Shenzhen 518120, China. (16) National Center for Nanoscience and Technology, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China. (17) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen 518083, China. (18) BGI Research, Shenzhen 518083, China. (19) BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China. (20) Zhejiang Hospital, Hangzhou 310013, China. Electronic address: yzq2229@163.com. (21) The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510655, China. Electronic address: wuxjian@mail.sysu.edu.cn. (22) National Center for Nanoscience and Technology, Beijing 100190, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGl-Shenzhen, Shenzhen 518120, China. Electronic address: zhaorf@nanoctr.cn. (23) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI, Shenzhen 518083, China. Electronic address: siqiliu@genomics.cn. (24) HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310000, China; BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen 518083, China. Electronic address: dongxuan@genomcs.cn.

Intratumoral three-cell-type clusters are a conserved feature of endogenous antitumor immunity

Damle and Carter et al. integrated spatial proteomics, tissue microarray, multiplex immunofluorescence imaging, and transcriptomics data from highly desmoplastic fibrolamellar carcinoma, and identified DC:Th:CTL three-cell-type clusters were present non-randomly in the TME. Immune triads were enriched for cDC1, mregDC, CXCL13+ Th, and GZMK+ effector CTL cells, showed upregulation of immune activation markers, and were present in treatment-naive murine and human PDAC. The density of APC:Th:CTL three-cell-type clusters in primary PDAC samples (n=467) correlated with intratumoral clonal T cell expansion and improved OS.

Contributed by Shishir Pant

Damle and Carter et al. integrated spatial proteomics, tissue microarray, multiplex immunofluorescence imaging, and transcriptomics data from highly desmoplastic fibrolamellar carcinoma, and identified DC:Th:CTL three-cell-type clusters were present non-randomly in the TME. Immune triads were enriched for cDC1, mregDC, CXCL13+ Th, and GZMK+ effector CTL cells, showed upregulation of immune activation markers, and were present in treatment-naive murine and human PDAC. The density of APC:Th:CTL three-cell-type clusters in primary PDAC samples (n=467) correlated with intratumoral clonal T cell expansion and improved OS.

Contributed by Shishir Pant

ABSTRACT: Effective antitumor immunity ultimately depends on the priming and activation of tumor-specific cytotoxic CD8+ T cells; however, the role of intratumoral cell-cell immune interactions remains incompletely understood. Recent work has revealed that the temporospatial colocalization of dendritic cells (DCs), helper T cells (Th), and cytotoxic T lymphocytes (CTL) within the tumor immune microenvironment following immune checkpoint blockade correlates with clinical response. Herein, we report the integration of more than one million spatially resolved single-cell profiles across six spatial proteomic and transcriptomic assays, which demonstrated that DC:Th:CTL three-cell-type clusters were common even in immunotherapy-nave and highly desmoplastic tumors, such as fibrolamellar carcinoma and pancreatic ductal adenocarcinoma (PDAC). We found that these immune triads were enriched for functionally important type 1 conventional DC, mature DCs enriched in immunoregulatory molecules (mregDC), CXCL13+ Th, and GZMK+ effector CTL phenotypes. Subsequent multiplex immunofluorescence imaging of more than 450 primary PDAC tumors showed that the density of antigen-presenting cell (APC):Th:CTL three-cell-type clusters was correlated with intratumoral T-cell clonal expansion and improved overall survival. These findings suggest that DC:Th:CTL triads are conserved across solid tumors and highlight the importance of intratumoral spatial niches in mediating endogenous antitumor immunity.

Author Info: (1) University of Washington, Seattle, WA, United States. (2) University of Washington, United States. (3) University of Washington, United States. (4) University of Washington, Se

Author Info: (1) University of Washington, Seattle, WA, United States. (2) University of Washington, United States. (3) University of Washington, United States. (4) University of Washington, Seattle, WA, United States. (5) University of Washington, Seattle, United States. (6) University of Washington, Seattle, Washington, United States. (7) Washington University in St. Louis, Saint Louis, MO, United States. (8) Washington University in St. Louis, Saint Louis, MO, United States. (9) University of Washington, Seattle, WA, United States. (10) University of Washington, Seattle, United States. (11) University of Washington, United States. (12) University of Washington, WA, United States. (13) Fred Hutchinson Cancer Center, Seattle, WA, United States. (14) Fred Hutchinson Cancer Center, Seattle, WA, United States. (15) Washington University in St. Louis, United States. (16) Washington University in St. Louis, St. Louis, MO, United States. (17) University of Washington, Seattle, WA, United States. (18) University of Washington, Seattle, WA, United States. (19) University of California, San Francisco, San Francisco, CA, United States. (20) Washington University in St. Louis, St. Louis, MO, United States. (21) University of Washington, Seattle, United States. (22) University of Washington, Seattle, WA, United States.

A modular mRNA platform for programmable induction of tumor-specific immunogenic cell death

Dong et al. encoded the ICD-inducing protein 4HB in LNPs optimized for tumor cell specificity and hepatocyte avoidance, which significantly improved tumor cargo expression compared to a clinical control LNP (>90% vs. ~25%). mRNA engineering via inclusion of UTRs complementary to non-tumor miRNAs (suppressive for liver) reduced off-target cargo distribution, markers of tissue injury, and body weight loss, without compromising tumor expression. The platform synergized with ICB in diverse “cold” tumor models via i.t. and i.v. routes, without systemic toxicity, enhancing infiltration of diverse immune cell types, including CD8+ T cells and macrophages.

Contributed by Morgan Janes

Dong et al. encoded the ICD-inducing protein 4HB in LNPs optimized for tumor cell specificity and hepatocyte avoidance, which significantly improved tumor cargo expression compared to a clinical control LNP (>90% vs. ~25%). mRNA engineering via inclusion of UTRs complementary to non-tumor miRNAs (suppressive for liver) reduced off-target cargo distribution, markers of tissue injury, and body weight loss, without compromising tumor expression. The platform synergized with ICB in diverse “cold” tumor models via i.t. and i.v. routes, without systemic toxicity, enhancing infiltration of diverse immune cell types, including CD8+ T cells and macrophages.

Contributed by Morgan Janes

ABSTRACT: Messenger RNA (mRNA) therapeutics hold great promise for oncology but their efficacy is limited by systemic off-target effects and immunosuppressive tumour microenvironments. Here we present TITUR, a tumour-customizable mRNA nanomedicine platform that integrates tumour-customizable ionizable lipids (TIs) and tumour-specific untranslated regions (TURs) to enhance tumour-selective mRNA delivery and expression. This dual-engineered approach enables the precise intratumoural expression of 4HB, an immunogenic cell death-inducing protein, while mitigating systemic toxicities. Using murine models of immunologically cold tumours, including melanoma and triple-negative breast cancer, TITUR-mediated 4HB delivery induced tumour-specific immunogenic cell death, remodelled the tumour microenvironment and enhanced immune cell infiltration. When combined with immune checkpoint inhibitors, 4HB TITUR suppressed primary and metastatic tumour growth, while also exhibiting vaccine-like properties by reducing tumour recurrence and eliciting systemic antitumour immunity. Furthermore, it demonstrated a superior safety profile compared with conventional mRNA delivery methods. Our data indicate that TITUR may serve as a versatile approach to address the limitations of current immunotherapies and support the development of personalized mRNA nanomedicines.

Author Info: (1) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (2) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (3) Leslie

Author Info: (1) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (2) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (3) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (4) Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. (5) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (6) Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. (7) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (8) Department of Chemistry, University of Toronto, Toronto, Ontario, Canada. (9) Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. (10) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (11) Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. (12) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (13) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (14) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. (15) Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. (16) Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. (17) Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. (18) Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. bw.li@utoronto.ca. Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. bw.li@utoronto.ca. Department of Chemistry, University of Toronto, Toronto, Ontario, Canada. bw.li@utoronto.ca. Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada. bw.li@utoronto.ca.

CD8+ T cell antitumor immunity via human iNKT-DC conjugates

Baiu et al. showed that CD4+ invariant natural killer T cells (iNKT) and autologous or allogeneic monocyte-derived DCs formed stable complexes, with enhanced expression of MHC I, 4-1BBL, OX40L, and IL-15Ra in DCs, and CD70 in iNKT cells. Complexes generated sustained DC signaling and created a platform for antigen-specific CD8+ T cell activation. In a xenograft model of B cell lymphoma, iNKT-DC induced T cell effector differentiation, reduced tumor burden, and remained effective at late disease stages that were resistant to ICB. Patient-derived DCs formed similar conjugates with allogeneic CD4+ iNKT cells and activated tumor antigen-specific CD8+ T cells.

Contributed by Shishir Pant

Baiu et al. showed that CD4+ invariant natural killer T cells (iNKT) and autologous or allogeneic monocyte-derived DCs formed stable complexes, with enhanced expression of MHC I, 4-1BBL, OX40L, and IL-15Ra in DCs, and CD70 in iNKT cells. Complexes generated sustained DC signaling and created a platform for antigen-specific CD8+ T cell activation. In a xenograft model of B cell lymphoma, iNKT-DC induced T cell effector differentiation, reduced tumor burden, and remained effective at late disease stages that were resistant to ICB. Patient-derived DCs formed similar conjugates with allogeneic CD4+ iNKT cells and activated tumor antigen-specific CD8+ T cells.

Contributed by Shishir Pant

ABSTRACT: Invariant Natural Killer T (iNKT) cells are a conserved T lymphocyte population capable of acting on dendritic cells (DCs) to potently amplify downstream immune responses. However, the processes underlying such iNKT adjuvancy remain poorly understood. Here, we showed that allogeneic human CD4+ iNKT cells form stably adhered bi-cellular complexes with monocyte-derived DCs that migrated together as pairs and showed extended DC calcium signaling. Compared to DCs treated with the synthetic adjuvant monophosphoryl lipid A (MPLA), DCs complexed with iNKT cells had elevated expression of MHC class I and multiple costimulatory molecules including 4-1BBL, OX40L, and IL-15R_, while the iNKT cells expressed CD70. Consistent with this distinctive co-stimulatory profile, iNKT-DC complexes were efficient activators of CD8+ T cells. Administering iNKT-DC complexes as a cellular immunotherapy in a xenograft model of aggressive human B cell lymphoma resulted in rapid reduction in tumor mass, antigen-specific B cell clearance, and transcriptional activation indicative of enhanced T cell proliferation and effector responses. iNKT-DC immunotherapy was effective at late stages of tumor progression that were refractory to immune checkpoint blockade immunotherapy, suggesting that the consortium of activating signals provided by iNKT-DC complexes rejuvenates exhausted antitumor immunity. Finally, allogeneic CD4+ iNKT cells formed similar complexes with monocyte-derived DCs from Head and Neck Cancer patients and promoted tumor antigen-dependent CD8+ T cell activation. These results show that monocyte-derived DCs paired with allogeneic CD4+ iNKT cells act as a potent antitumor cellular immunotherapy that activates antigen-specific CD8+ T cell immunity.

Author Info: (1) University of Wisconsin-Madison, Madison, WI, United States. (2) University of Wisconsin-Madison, Madison, WI, United States. (3) University of Wisconsin-Madison, Madison, WI,

Author Info: (1) University of Wisconsin-Madison, Madison, WI, United States. (2) University of Wisconsin-Madison, Madison, WI, United States. (3) University of Wisconsin-Madison, Madison, WI, United States. (4) University of Wisconsin-Madison, Madison, WI, United States. (5) University of Wisconsin-Madison, Madison, WI, United States. (6) University of Wisconsin-Madison, United States. (7) University of Wisconsin-Madison, Madison, WI, United States. (8) University of Wisconsin-Madison, Madison, WI, United States. (9) (10) University of Wisconsin-Madison, Madison, WI, United States.

Ms4a7 expression in cDC1s determines cross-­presentation and antitumor immunity

Focused on key TME regulators that control cDC1 cross-presentation, Xie et al. found that cDC1-expressed Ms4a7 was critical for effective CD8+ T cell-mediated antitumor immunity in mouse models. Tumor antigen induced Ms4a7 expression and NF-κB activation in cDC1s and tumor dLNs, which was required to cross-prime antigen-specific CD8+ T cells. Ms4a7-/- cDC1s showed reduced migration to dLNs, made less IL-­12, IL-­18, and IL-­27 in the TME, and failed to prime CD8+ T cell activation, resulting in diminished antitumor immunity. In human cancers, Ms4a7 was expressed in a subset of cDC1s, enriched in dLNs, and correlated with patient survival.

Contributed by Katherine Turner

Focused on key TME regulators that control cDC1 cross-presentation, Xie et al. found that cDC1-expressed Ms4a7 was critical for effective CD8+ T cell-mediated antitumor immunity in mouse models. Tumor antigen induced Ms4a7 expression and NF-κB activation in cDC1s and tumor dLNs, which was required to cross-prime antigen-specific CD8+ T cells. Ms4a7-/- cDC1s showed reduced migration to dLNs, made less IL-­12, IL-­18, and IL-­27 in the TME, and failed to prime CD8+ T cell activation, resulting in diminished antitumor immunity. In human cancers, Ms4a7 was expressed in a subset of cDC1s, enriched in dLNs, and correlated with patient survival.

Contributed by Katherine Turner

ABSTRACT: Conventional type 1 dendritic cells (cDC1s) capture antigens in peripheral tissues and migrate to draining lymph nodes (dLNs) to prime antigen-specific CD8(+) T cells. How tumor antigens are processed to activate CD8(+) T cell immunity is not well understood. In this work, we show that Ms4a7 is up-regulated in cDC1s after tumor antigen uptake or exposure to exogenous stimuli and is required for their cross-priming ability. Although Ms4a7(-/-) mice showed normal cDC1 development and turnover, they failed to prime antigen-specific CD8(+) T cells following infection or tumor development. In human cancers, MS4A7 was expressed in a subset of cDC1s, preferentially enriched in dLNs, and correlated with patient survival. Our findings suggest a critical role for Ms4a7 in cDC1-mediated cross-presentation and antitumor CD8(+) T cell responses.

Author Info: (1) Westlake University School of Medicine, Hangzhou, Zhejiang, China. Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. Tsinghua-Peking Center

Author Info: (1) Westlake University School of Medicine, Hangzhou, Zhejiang, China. Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China. (2) Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China. (3) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (4) Westlake University School of Medicine, Hangzhou, Zhejiang, China. Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (5) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (6) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China. (7) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (8) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (9) Westlake University School of Medicine, Hangzhou, Zhejiang, China. Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (10) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (11) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (12) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (13) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (14) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China. (15) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (16) Westlake University School of Medicine, Hangzhou, Zhejiang, China. (17) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (18) Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China. (19) Westlake University School of Medicine, Hangzhou, Zhejiang, China.

‘‘Cancer-treating-cancer’’ strategy- Entrapping chemically engineered dying cancer cells in immunotherapeutic hydrogel against tumor recurrence

Spotlight 

Zhang et al employed cytotoxic B-elemene (ELE)-treated dying cancer cells (DCCs) as an immunogenic cell source within a sprayable fibrin hydrogel (with additional ELE). In a 4T1 orthotopic partial resection model, the gel controlled primary and untreated distal tumor growth, increasing the M1:M2 macrophage ratio, and activated CD103+ DCs. The gel increased T cell frequency in the tumor, lymph nodes, and spleen, Tcm polarization, and stimulatory cytokines. Proteomics profiling of tumor fluid and plasma revealed differential expression of clinically prognostic proteins and pathways related to apoptosis, immune function, and metastasis.

Contributed by Morgan Janes

Zhang et al employed cytotoxic B-elemene (ELE)-treated dying cancer cells (DCCs) as an immunogenic cell source within a sprayable fibrin hydrogel (with additional ELE). In a 4T1 orthotopic partial resection model, the gel controlled primary and untreated distal tumor growth, increasing the M1:M2 macrophage ratio, and activated CD103+ DCs. The gel increased T cell frequency in the tumor, lymph nodes, and spleen, Tcm polarization, and stimulatory cytokines. Proteomics profiling of tumor fluid and plasma revealed differential expression of clinically prognostic proteins and pathways related to apoptosis, immune function, and metastasis.

Contributed by Morgan Janes

ABSTRACT: Postsurgical tumor recurrence remains a major challenge, primarily driven by the resurgence of residual microtumors at surgical margins. The tumor microenvironment (TME) in these regions plays a decisive role in treatment outcomes. Here, we present an in situ sprayed fibrin hydrogel system that integrates chemically engineered homologous dying cancer cells (DCCs) as a sustained antigen reservoir with the anticancer agent β-elemene (ELE) to enhance anti-tumor immune responses and suppress local tumor recurrence. This immunotherapeutic hydrogel (DCCs@ELE@Gel) modulates the TME by promoting a favorable M1/M2 tumor-associated macrophage balance, facilitating dendritic cell maturation, and enhancing the cross-priming of cytotoxic T cells, collectively preventing tumor regrowth. Additionally, comprehensive proteomic analysis reveals key mechanisms linking the chemo-immunotherapeutic hydrogel to tumor recurrence suppression. Our findings introduce an approach that leverages engineered tumor cells within a hydrogel matrix for improved cancer immunotherapy, offering a versatile strategy for postsurgical tumor management.

Author Info: 1- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Ey

Author Info: 1- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China 2- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China 3- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA 4- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China 5- Lead contact

Trilaciclib triggers a neutrophil-related immune response and sensitizes non-small cell lung cancer to anti-PD-1 therapy Spotlight 

Gao, He, and Wang et al. show that Trilaciclib (CDK4/6 inhibitor) inhibits NSCLC progression in immunocompetent, but not immunodeficient, mice bearing s.c. LLC and CMT167 tumors. Trilaciclib induced tumor cell senescence and SASP in a cGAS-STING-dependent manner, promoted infiltration and activation of anti-tumor CD177+ neutrophils in the tumor, and activated CD8+ T cells with enhanced effector and cytotoxic function, partly via a cytokine-dependent priming. Trilaciclib enhanced the efficacy of immune checkpoint inhibitors and reduced tumor growth, without observable toxicity, in an LLC tumor model and patient derived organoids.

Contributed by Shishir Pant

Gao, He, and Wang et al. show that Trilaciclib (CDK4/6 inhibitor) inhibits NSCLC progression in immunocompetent, but not immunodeficient, mice bearing s.c. LLC and CMT167 tumors. Trilaciclib induced tumor cell senescence and SASP in a cGAS-STING-dependent manner, promoted infiltration and activation of anti-tumor CD177+ neutrophils in the tumor, and activated CD8+ T cells with enhanced effector and cytotoxic function, partly via a cytokine-dependent priming. Trilaciclib enhanced the efficacy of immune checkpoint inhibitors and reduced tumor growth, without observable toxicity, in an LLC tumor model and patient derived organoids.

Contributed by Shishir Pant

ABSTRACT: Immunotherapy-based combination approaches have improved treatment efficacy in advanced non-small cell lung cancer (NSCLC), but progressive disease remains a challenge. Trilaciclib is a cyclin-dependent kinase 4/6 inhibitor approved for myelopreservation in extensive-stage small cell lung cancer (ES-SCLC). Our results demonstrate that trilaciclib has antitumor potential in NSCLC without significant toxicity. It reprograms the tumor immune microenvironment by primarily increasing antitumor neutrophils and CD8(+) T cells. Trilaciclib induces tumor cell senescence and the senescence-associated secretory phenotype in a cGAS-STING-dependent manner, which further facilitates the infiltration and activation of CD177(+) neutrophils with anti-tumor properties. These neutrophils enhance CD8(+) effector T cell activation and promote antitumor immunity. Additionally, activated CD8(+) T cells recruit and activate neutrophils, forming a positive feedback loop. Combining trilaciclib with anti-PD-1 antibodies presents a promising strategy for NSCLC treatment.

Author Info: (1) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Department of Thoracic Oncology, Lung Cancer Diagnosis and Tr

Author Info: (1) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Department of Thoracic Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100053, China. (2) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. (3) National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China. (4) National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China. (5) National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Department of Endoscopy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China. (6) National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China. (7) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. (8) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. (9) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Department of Thoracic Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. (10) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. (11) Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China. (12) National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China. (13) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. (14) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. (15) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. Electronic address: zhiyongliu@tjmuch.com. (16) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. Electronic address: chenpeng@tjmuch.com. (17) Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; National Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China. Electronic address: guohua@tjmuch.com.

Developing a therapeutic elastase that stimulates anti-tumor immunity by selectively killing cancer cells Featured  

Gujar and Cui et al. evaluated N17350, a next-generation therapeutic elastase, and found that it selectively induced immunogenic cell death in cancer cells (based on their increased expression of histone H1.0 and H1.2 proteins), driving tumor regression and inducing systemic antitumor immunity across a wide range of preclinical models. N17350 remained effective with repeated dosing and in resistant cancers, and synergized with immunotherapies, positioning it as a strong candidate for first-in-human clinical evaluation.

Gujar and Cui et al. evaluated N17350, a next-generation therapeutic elastase, and found that it selectively induced immunogenic cell death in cancer cells (based on their increased expression of histone H1.0 and H1.2 proteins), driving tumor regression and inducing systemic antitumor immunity across a wide range of preclinical models. N17350 remained effective with repeated dosing and in resistant cancers, and synergized with immunotherapies, positioning it as a strong candidate for first-in-human clinical evaluation.

ABSTRACT: Recent clinical studies highlight the effectiveness of combining cytotoxic agents with immunotherapies, emphasizing the need for next-generation treatments that integrate both therapeutic approaches. Here, we use 30 cancer cell lines, 15 tumor models, and 45 patient samples to develop N17350, a therapeutic elastase that targets the "neutrophil elastase pathway" to induce tumor regression and stimulate anti-tumor immunity. N17350 leverages linker histone H1.0 and H1.2, proteins elevated in many cancers, to trigger immunogenic cancer cell death while preserving immune cells. Intra-tumoral N17350 administration induces rapid, genotype-independent tumor regression, triggering CD8(+) T cell activation to promote durable responses and enable checkpoint inhibitor efficacy in refractory models. N17350 maintains potency with repeated dosing and across diverse treatment histories, including resistance to chemotherapies and checkpoint inhibitors. These findings support the advancement of N17350 to first-in-human clinical trials as a cytotoxic agent designed to stimulate anti-tumor immunity by selectively killing cancer cells.

Author Info: (1) Onchilles Pharma Inc., San Diego, CA, USA. (2) Onchilles Pharma Inc., San Diego, CA, USA. (3) Onchilles Pharma Inc., San Diego, CA, USA. (4) Onchilles Pharma Inc., San Diego, C

Author Info: (1) Onchilles Pharma Inc., San Diego, CA, USA. (2) Onchilles Pharma Inc., San Diego, CA, USA. (3) Onchilles Pharma Inc., San Diego, CA, USA. (4) Onchilles Pharma Inc., San Diego, CA, USA. (5) South Coast Gynecologic Oncology Inc, San Diego, CA, USA. (6) Helen Diller Family Comprehensive Cancer Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA. (7) The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust National Institute of Health Research Biomedical Research Centre, London, UK. (8) Onchilles Pharma Inc., San Diego, CA, USA. (9) Onchilles Pharma Inc., San Diego, CA, USA. Electronic address: lbecker@onchillespharma.com.

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.