Journal Articles

Combination of pembrolizumab and radiotherapy induces systemic antitumor immune responses in immunologically cold non-small cell lung cancer

The abscopal effects of radiation may sensitize immunologically cold tumors to immune checkpoint inhibition. We investigated the immunostimulatory effects of radiotherapy leveraging multiomic analyses of serial tissue and blood biospecimens (n_=_293) from a phase 2 clinical trial of stereotactic body radiation therapy (SBRT) followed by pembrolizumab in metastatic non-small cell lung cancer ( NCT02492568 ). Participants with immunologically cold tumors (low tumor mutation burden, null programmed death ligand 1 expression or Wnt pathway mutations) had significantly longer progression-free survival in the SBRT arm. Induction of interferon-_, interferon-_ and antigen processing and presentation gene sets was significantly enriched after SBRT in nonirradiated tumor sites. Significant on-therapy expansions of new and pre-existing T cell clones in both the tumor (abscopal) and the blood (systemic) compartments were noted alongside clonal neoantigen-reactive autologous T cell responses in participants with long-term survival after radioimmunotherapy. These findings support the systemic immunomodulatory and antitumor effects of radioimmunotherapy and may open a therapeutic window of opportunity to overcome immunotherapy resistance.

Author Info: (1) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (2) Netherlands Cancer Institute, Amsterdam, The Netherlands. (3

Author Info: (1) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (2) Netherlands Cancer Institute, Amsterdam, The Netherlands. (3) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (4) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (5) Netherlands Cancer Institute, Amsterdam, The Netherlands. (6) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. The Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (7) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (8) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (9) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (10) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (11) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA. (12) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (13) Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands. (14) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (15) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. The Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA. (16) Netherlands Cancer Institute, Amsterdam, The Netherlands. (17) The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. vanagno1@jhmi.edu. The Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA. vanagno1@jhmi.edu.

De novo-designed pMHC binders facilitate T cell-mediated cytotoxicity toward cancer cells

The recognition of intracellular antigens by CD8(+) T cells through T cell receptors (TCRs) is central for adaptive immunity against infections and cancer. However, the identification of TCRs from patient material remains complex. We present a rapid de novo minibinder (miBd) design platform leveraging state-of-the-art generative models to engineer miBds targeting the cancer-associated peptide-bound major histocompatibility complex (pMHC) SLLMWITQC/HLA-A*02:01 (NY-ESO-1). Incorporating in silico cross-panning enabled computational prescreening of specificity, and molecular dynamics simulations allowed for improved predictability of in vitro success. We identified a high-affinity NY-ESO-1 binder and confirmed its structure using cryo-electron microscopy, which, when incorporated in a chimeric antigen receptor, induced killing of NY-ESO-1(+) melanoma cells. We further designed and validated binders to a neoantigen pMHC complex, RVTDESILSY/HLA-A*01:01, with unknown structure, demonstrating the potential for precision immunotherapy.

Author Info: (1) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (2) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kon

Author Info: (1) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (2) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (3) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (4) Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA. (5) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (6) Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA. (7) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (8) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (9) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (10) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (11) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (12) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (13) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (14) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (15) Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA. (16) Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA. (17) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (18) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (19) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (20) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (21) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (22) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (23) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark. (24) Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA. (25) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (26) Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark. (27) Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark.

De novo design and structure of a peptide-centric TCR mimic binding module

T cell receptor (TCR) mimics offer a promising platform for tumor-specific targeting of peptide-major histocompatibility complex (pMHC) in cancer immunotherapy. In this study, we designed a de novo _-helical TCR mimic (TCRm) specific for the NY-ESO-1 peptide presented by human leukocyte antigen (HLA)-A*02, achieving high on-target specificity with nanomolar affinity (dissociation constant K(d) = 9.5 nM). The structure of the TCRm-pMHC complex at 2.05- resolution revealed a rigid TCR-like docking mode with an unusual degree of focus on the up-facing NY-ESO-1 side chains, suggesting the potential for reduced off-target reactivity. Indeed, a structure-informed in silico screen of 14,363 HLA-A*02 peptides correctly predicted two off-target peptides, yet our TCRm maintained peptide selectivity and cytotoxicity as a T cell engager. These results represent a path for precision targeting of tumor antigens with peptide-focused _-helical TCR mimics.

Author Info: (1) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. Program in Immunology, Stanford University School of Medicine, Stanf

Author Info: (1) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA. (2) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. (3) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. (4) Department of Computer Science, Stanford University, Stanford, CA, USA. (5) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. (6) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. (7) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. (8) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. (9) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA. (10) Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA. Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.

Design of high-specificity binders for peptide-MHC-I complexes

Class I major histocompatibility complex (MHC-I) molecules present peptides derived from intracellular antigens on the cell surface for immune surveillance. Proteins that recognize peptide-MHC-I (pMHCI) complexes with specificity for diseased cells could have considerable therapeutic utility. Specificity requires recognition of outward-facing amino acid residues within the disease-associated peptide as well as avoidance of extensive contacts with ubiquitously expressed MHC. We used RFdiffusion to design pMHCI-binding proteins that make extensive contacts with the peptide and identified specific binders for 11 target pMHCs starting from either experimental or predicted pMHCI structures. Upon incorporation into chimeric antigen receptors, designs for eight targets conferred peptide-specific T cell activation. Our approach should have broad utility for both protein- and cell-based pMHCI targeting.

Author Info: (1) Department of Biochemistry, University of Washington, Seattle, WA, USA. Institute for Protein Design, University of Washington, Seattle, WA, USA. (2) Department of Biochemistry

Author Info: (1) Department of Biochemistry, University of Washington, Seattle, WA, USA. Institute for Protein Design, University of Washington, Seattle, WA, USA. (2) Department of Biochemistry, University of Washington, Seattle, WA, USA. Institute for Protein Design, University of Washington, Seattle, WA, USA. (3) Department of Biochemistry, University of Washington, Seattle, WA, USA. Institute for Protein Design, University of Washington, Seattle, WA, USA. Bioengineering Graduate Program, University of Washington, Seattle, WA, USA. (4) Department of Biochemistry, University of Washington, Seattle, WA, USA. Institute for Protein Design, University of Washington, Seattle, WA, USA. Bioengineering Graduate Program, University of Washington, Seattle, WA, USA. (5) Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. (6) Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. (7) Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA, USA. (8) Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA. Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA. (9) Department of Biochemistry, University of Washington, Seattle, WA, USA. Institute for Protein Design, University of Washington, Seattle, WA, USA. (10) Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA, USA. (11) Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA. (12) Department of Biochemistry, University of Washington, Seattle, WA, USA. Institute for Protein Design, University of Washington, Seattle, WA, USA. (13) Department of Biochemistry, University of Washington, Seattle, WA, USA. Institute for Protein Design, University of Washington, Seattle, WA, USA. (14) Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA. Department of Physics, University of Washington, Seattle, WA, USA. Department of Applied Mathematics, University of Washington, Seattle, WA, USA. Fred Hutchinson Cancer Center, Seattle, WA, USA. (15) Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. Weill Cornell Medicine, New York, NY, USA. (16) Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA, USA. Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA. (17) Department of Biochemistry, University of Washington, Seattle, WA, USA. Institute for Protein Design, University of Washington, Seattle, WA, USA. Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.

Microbiota-driven antitumour immunity mediated by dendritic cell migration Spotlight 

Lin, Fukuoka, and Koyama et al. focused on how gut microbiota influence the clinical efficacy of ICB. A newly identified bacterial strain named YB328 (genus hominenteromicrobium), was significantly elevated in feces of patients who responded to PD-1 inhibition. Mechanistically, YB328 promoted antitumor efficacy by activating tumor-specific CD103+CD11b- conventional DCs in the gut via multiple TLRs, promoting their migration and in situ activation of a broader repertoire of PD1+CD8+ T cells in the dLNs and TME. YB328 enhanced ICB efficacy in multiple tumor models, and levels correlated with PFS in various cancer types.

Contributed by Katherine Turner

Lin, Fukuoka, and Koyama et al. focused on how gut microbiota influence the clinical efficacy of ICB. A newly identified bacterial strain named YB328 (genus hominenteromicrobium), was significantly elevated in feces of patients who responded to PD-1 inhibition. Mechanistically, YB328 promoted antitumor efficacy by activating tumor-specific CD103+CD11b- conventional DCs in the gut via multiple TLRs, promoting their migration and in situ activation of a broader repertoire of PD1+CD8+ T cells in the dLNs and TME. YB328 enhanced ICB efficacy in multiple tumor models, and levels correlated with PFS in various cancer types.

Contributed by Katherine Turner

ABSTRACT: Gut microbiota influence the antitumour efficacy of immune checkpoint blockade(1-6), but the mechanisms of action have not been fully elucidated. Here, we show that a new strain of the bacterial genus Hominenteromicrobium (designated YB328) isolated from the faeces of patients who responded to programmed cell death_1 (PD-1) blockade augmented antitumour responses in mice. YB328 activated tumour-specific CD8(+) T_cells through the stimulation of CD103(+)CD11b(-) conventional dendritic cells (cDCs), which, following exposure in the gut, migrated to the tumour microenvironment. Mice showed improved antitumour efficacy of PD-1 blockade when treated with faecal transplants from non-responder patients supplemented with YB238. This result suggests that YB328 could function in a dominant manner. YB328-activated CD103(+)CD11b(-) cDCs showed prolonged engagement with tumour-specific CD8(+) T_cells and promoted PD-1 expression in these cells. Moreover, YB238-augmented antitumour efficacy of PD-1 blockade treatment was observed in multiple mouse models of cancer. Patients with elevated YB328 abundance had increased infiltration of CD103(+)CD11b(-) cDCs in tumours and had a favourable response to PD-1 blockade therapy in various cancer types. We propose that gut microbiota enhance antitumour immunity by accelerating the maturation and migration of CD103(+)CD11b(-) cDCs to increase the number of CD8(+) T_cells that respond to diverse tumour antigens.

Author Info: (1) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan.

Author Info: (1) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan. (2) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. (3) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan. (4) Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan. (5) Molecular Biosystems Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan. (6) Benno Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science Technology and Innovation Laboratory, Saitama, Japan. (7) Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan. (8) Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan. (9) Molecular Biosystems Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan. (10) Biomanufacturing Process Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan. (11) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. (12) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. Department of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan. (13) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan. (14) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. (15) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. (16) Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan. (17) Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan. (18) Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan. (19) Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan. (20) Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan. (21) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. (22) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. (23) Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Chiba, Japan. (24) Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Chiba, Japan. (25) Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba, Japan. (26) Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Chiba, Japan. Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan. (27) Department of Head and Neck Oncology and Innovative Treatment, Graduate School of Medicine, Kyoto University, Kyoto, Japan. (28) Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan. (29) Department of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan. (30) Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan. (31) Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan. (32) Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan. (33) Molecular Biosystems Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan. (34) Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan. (35) Benno Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science Technology and Innovation Laboratory, Saitama, Japan. benno828@bigm.or.jp. Benno Institute for Gut Microflora (BIGM), Saitama Industrial Technology Center, Saitama, Japan. benno828@bigm.or.jp. (36) Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan. hnishika@ncc.go.jp. Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan. hnishika@ncc.go.jp. Division of Cancer Immune Multicellular System Regulation, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan. hnishika@ncc.go.jp. Kindai University Faculty of Medicine, Osaka, Japan. hnishika@ncc.go.jp.

Bispecific killer cell engager-secreting CAR-T cells redirect natural killer specificity to enhance antitumour responses Spotlight 

Testing various combinations, Fan et al. found that the administration of bispecific killer cell-engager (BiKE)-secreting CAR T cells alongside weekly injections of NK cells was optimal for achieving long-term control in murine hematologic tumor models. In a solid tumor model, NK cells co-administred with BiKE+ CAR T cells showed tumor parenchyma infiltration, whereas NK cells co-administred with BiKE- CAR T cells were primarily found in the peritumoral connective tissue. The simultaneous expression of a CD19-targeting CAR and EGFR-targeting BIKEs in T cell led to complete eradication of heterogeneous EGFR+CD19- and EGFR-CD19+ tumor cells in vivo.

Contributed by Ute Burkhardt

Testing various combinations, Fan et al. found that the administration of bispecific killer cell-engager (BiKE)-secreting CAR T cells alongside weekly injections of NK cells was optimal for achieving long-term control in murine hematologic tumor models. In a solid tumor model, NK cells co-administred with BiKE+ CAR T cells showed tumor parenchyma infiltration, whereas NK cells co-administred with BiKE- CAR T cells were primarily found in the peritumoral connective tissue. The simultaneous expression of a CD19-targeting CAR and EGFR-targeting BIKEs in T cell led to complete eradication of heterogeneous EGFR+CD19- and EGFR-CD19+ tumor cells in vivo.

Contributed by Ute Burkhardt

ABSTRACT: T cells and natural killer (NK) cells collaborate to maintain immune homeostasis. Current cancer immunotherapies predominantly rely on the individual application of these cells. Here we use bicistronic vectors to co-express chimeric antigen receptors (CARs) and secreted immune cell engagers (ICEs), leveraging the combined therapeutic potential of both effector cell types. After in vitro validation of immune cell engager secretion and function, various combinatorial approaches are systematically compared in mouse models, identifying a highly effective combination of bispecific killer cell engager (BiKE)-secreting CAR-T cells and NK cells. Beyond a simple combination of conventional CAR-T cells and NK cells, this strategy demonstrates superior efficacy in CD19(+) B cell leukaemia and lymphoma and EGFR(+) solid tumour models while reducing the dosage dependence on CAR-T cells. Moreover, CAR-T cells and BiKEs targeting distinct antigens exhibit suppression of tumour cells with heterogeneous antigen expression. These findings indicate that combining BiKE-secreting CAR-T cells and NK cells offers a promising strategy to combat tumour antigen heterogeneity and immune evasion.

Author Info: (1) Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. Liangzhu Laboratory, Zhejiang University, Hangzhou,

Author Info: (1) Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. Liangzhu Laboratory, Zhejiang University, Hangzhou, China. (2) Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. Liangzhu Laboratory, Zhejiang University, Hangzhou, China. Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China. Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China. (3) Liangzhu Laboratory, Zhejiang University, Hangzhou, China. Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China. Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China. (4) Liangzhu Laboratory, Zhejiang University, Hangzhou, China. Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China. Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China. (5) Liangzhu Laboratory, Zhejiang University, Hangzhou, China. Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China. Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China. (6) Liangzhu Laboratory, Zhejiang University, Hangzhou, China. Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China. Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China. (7) School of Public Health, Zhejiang University School of Medicine, Hangzhou, China. (8) School of Public Health, Zhejiang University School of Medicine, Hangzhou, China. (9) Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, New York, NY, USA. (10) Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. huanghe@zju.edu.cn. Liangzhu Laboratory, Zhejiang University, Hangzhou, China. huanghe@zju.edu.cn. Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China. huanghe@zju.edu.cn. (11) Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. sunj4@zju.edu.cn. Liangzhu Laboratory, Zhejiang University, Hangzhou, China. sunj4@zju.edu.cn. Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China. sunj4@zju.edu.cn. Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China. sunj4@zju.edu.cn.

Tissue origin and virus specificity shape human CD8+ T cell cytotoxicity

Spotlight 

Niessl et al. found that blood-derived memory CD8+ T cells exhibited greater basal cytotoxic molecule expression than T cells from matched tissue, while resident memory cells exhibited the weakest cytotoxicity profile. Blood and tissue (tonsillar) effector memory CD8+ T cells also exhibited differential responses to TCR activation and cytokine stimulation (TGFβ/IL-15). Antigen specificity played a significant role in site residence and phenotype, wherein chronic virus-associated T cells exhibited greater cytotoxicity. While expression of diverse granzymes was considered a cytotoxic hallmark in blood T cells, only granzyme B was found to contribute to cell killing.

Contributed by Morgan Janes

Niessl et al. found that blood-derived memory CD8+ T cells exhibited greater basal cytotoxic molecule expression than T cells from matched tissue, while resident memory cells exhibited the weakest cytotoxicity profile. Blood and tissue (tonsillar) effector memory CD8+ T cells also exhibited differential responses to TCR activation and cytokine stimulation (TGFβ/IL-15). Antigen specificity played a significant role in site residence and phenotype, wherein chronic virus-associated T cells exhibited greater cytotoxicity. While expression of diverse granzymes was considered a cytotoxic hallmark in blood T cells, only granzyme B was found to contribute to cell killing.

Contributed by Morgan Janes

ABSTRACT: CD8(+) T cells are classically defined by cytotoxic activity, but it has remained unclear whether cytotoxic programs are compartmentalized across tissues and memory subsets. Here, we established a human organ donor cohort and found that expression of conventional cytotoxic molecules-granulysin, perforin, and granzyme B-was most prominent among circulating memory CD8(+) T cells and decreased progressively with tissue residency, inversely mirroring the expression of CD69 and CD103. Other cytotoxic molecules, including granzymes A, H, K, and M, were variably expressed across tissues, and memory CD8(+) T cells targeting persistent viruses expressed multiple granzymes coordinately. In an in vitro tonsil system, transforming growth factor-_ induced discordant regulation of cytotoxic molecules and CD103. Combined with interleukin-15, this circuitry modulated proliferation and the acquisition of redirected killing activity via perforin and granzyme B. Our findings suggest that human memory CD8(+) T cell cytotoxicity is intricately regulated by environmental cues reflecting tissue location and antigen specificity.

Author Info: (1) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (2) Center for Infectious Medicine, Department of Medicine Huddinge,

Author Info: (1) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (2) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (3) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (4) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (5) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (6) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (7) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (8) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (9) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (10) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (11) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (12) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (13) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (14) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (15) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (16) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (17) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (18) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (19) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (20) Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (21) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (22) Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (23) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (24) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (25) Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK. (26) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (27) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (28) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (29) Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK. Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK. (30) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (31) Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (32) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (33) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden. (34) ME Transplantation, Karolinska University Hospital Huddinge, Stockholm, Sweden. Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden. (35) Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.

Sensitization of tumours to immunotherapy by boosting early type-I interferon responses enables epitope spreading Spotlight 

Qdaisat, Wummer, and Stover et al. demonstrated that early type-I interferon responses restored a defective damage response in poorly immunogenic tumors to enable epitope spreading and sensitivity to ICB. ICB response was transferrable to resistant models due to epitope spread against poorly immunogenic tumor antigens in an IFNAR1-dependent manner. Systemic administration of lipid particles loaded with RNA coding for tumor-unspecific antigens enhanced interferon responses, induced epitope spreading, and reprogrammed the TME from myeloid suppressive to pro-inflammatory, favoring effector T cells for sustained immune response in poorly immunogenic models.

Contributed by Shishir Pant

Qdaisat, Wummer, and Stover et al. demonstrated that early type-I interferon responses restored a defective damage response in poorly immunogenic tumors to enable epitope spreading and sensitivity to ICB. ICB response was transferrable to resistant models due to epitope spread against poorly immunogenic tumor antigens in an IFNAR1-dependent manner. Systemic administration of lipid particles loaded with RNA coding for tumor-unspecific antigens enhanced interferon responses, induced epitope spreading, and reprogrammed the TME from myeloid suppressive to pro-inflammatory, favoring effector T cells for sustained immune response in poorly immunogenic models.

Contributed by Shishir Pant

ABSTRACT: The success of cancer immunotherapies is predicated on the targeting of highly expressed neoepitopes, which preferentially favours malignancies with high mutational burden. Here we show that early responses by type-I interferons mediate the success of immune checkpoint inhibitors as well as epitope spreading in poorly immunogenic tumours and that these interferon responses can be enhanced via systemic administration of lipid particles loaded with RNA coding for tumour-unspecific antigens. In mice, the immune responses of tumours sensitive to checkpoint inhibitors were transferable to resistant tumours and resulted in heightened immunity with antigenic spreading that protected the animals from tumour rechallenge. Our findings show that the resistance of tumours to immunotherapy is dictated by the absence of a damage response, which can be restored by boosting early type-I interferon responses to enable epitope spreading and self-amplifying responses in treatment-refractory tumours.

Author Info: (1) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. Univer

Author Info: (1) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. University of Florida Genetics Institute, University of Florida, Gainesville, FL, USA. (2) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (3) Department of Pediatrics, Division of Pediatric Hematology-Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA. (4) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (5) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (6) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (7) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (8) Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (9) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (10) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (11) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (12) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (13) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (14) Department of Pediatrics, Division of Pediatric Hematology-Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA. (15) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (16) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (17) Department of Pediatrics, Division of Pediatric Hematology-Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA. (18) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (19) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (20) Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, USA. (21) Department of Medicine, Division of Hematology and Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA. (22) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (23) Department of Pediatrics, Division of Pediatric Hematology-Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA. (24) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (25) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (26) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (27) Department of Pediatrics, Division of Pediatric Hematology-Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA. (28) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (29) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (30) College of Veterinary Medicine, University of Florida, Gainesville, FL, USA. (31) Department of Pathology, University of Florida, Gainesville, FL, USA. (32) Center of Immunotherapy and Precision Immuno-Oncology/Head and Neck Institute, Cleveland Clinic, Cleveland, OH, USA. (33) Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, USA. (34) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (35) Department of Pediatrics, Division of Pediatric Hematology-Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA. (36) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (37) Department of Biostatistics, University of Florida, Gainesville, FL, USA. (38) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (39) Department of Pediatrics, Division of Pediatric Hematology-Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA. (40) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (41) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. Elias.Sayour@neurosurgery.ufl.edu. Department of Pediatrics, Division of Pediatric Hematology-Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA. Elias.Sayour@neurosurgery.ufl.edu.

Systemic administration of an RNA binding and cell-penetrating antibody targets therapeutic RNA to multiple mouse models of cancer Spotlight 

Quijano, Martinez-Saucedo, and Ianniello et al. showed that the mAb TMAB3 noncovalently bound to 3p-hpRNA to form stable antibody/RNA complexes that delivered 3p-hpRNA specifically to premalignant and malignant cells. Intravenously administered TMAB3/ 3p-hpRNA complexes enhanced survival and suppressed tumor growth in orthotopic pancreatic cancer and medulloblastoma mouse models. Single-cell RNAseq showed that TMAB3/3p-hpRNA treatment promoted cytotoxic T cell infiltration and RIG-I activation. T cell activation mediated by TMAB3/3p-hpRNA was dependent on ENT2 and RIG-I expression on PDAC cells.

Contributed by Shishir Pant

Quijano, Martinez-Saucedo, and Ianniello et al. showed that the mAb TMAB3 noncovalently bound to 3p-hpRNA to form stable antibody/RNA complexes that delivered 3p-hpRNA specifically to premalignant and malignant cells. Intravenously administered TMAB3/ 3p-hpRNA complexes enhanced survival and suppressed tumor growth in orthotopic pancreatic cancer and medulloblastoma mouse models. Single-cell RNAseq showed that TMAB3/3p-hpRNA treatment promoted cytotoxic T cell infiltration and RIG-I activation. T cell activation mediated by TMAB3/3p-hpRNA was dependent on ENT2 and RIG-I expression on PDAC cells.

Contributed by Shishir Pant

ABSTRACT: There is intense interest in the advancement of RNAs as rationally designed therapeutic agents, especially in oncology, where a major focus is to use RNAs to stimulate pattern recognition receptors to leverage innate immune responses. However, the inability to selectively deliver therapeutic RNAs within target cells after intravenous administration now hinders the development of this type of treatment for cancer and other disorders. Here, we found that a tumor-targeting, cell-penetrating, and RNA binding monoclonal antibody, TMAB3, can form stable, noncovalent antibody/RNA complexes of a discrete size that mediate highly specific and functional delivery of RNAs into tumors. Using 3p-hpRNA, an agonist of the pattern recognition receptor retinoic acid-inducible gene-I (RIG-I), we observed robust antitumor efficacy of systemically administered TMAB3/3p-hpRNA complexes in mouse models of pancreatic cancer, medulloblastoma, and melanoma. In the KPC syngeneic, orthotopic pancreatic cancer model in immunocompetent mice, treatment with TMAB3/3p-hpRNA tripled animal survival, decreased tumor growth, and specifically targeted malignant cells, with a 1500-fold difference in RNA delivery into tumor cells versus nonmalignant cells within the tumor mass. Single-cell RNA sequencing (scRNA-seq) and flow cytometry demonstrated that TMAB3/3p-hpRNA treatment elicited a potent antitumoral immune response characterized by RIG-I activation and increased infiltration and activity of cytotoxic T cells. These studies established that TMAB3/RNA complexes can deliver RNA payloads specifically to hard-to-treat tumor cells to achieve antitumor efficacy, providing an antibody-based platform to advance the study of RNA therapies for the treatment of patients with cancer.

Author Info: (1) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. Department of Genetics, Yale University School of Medicine, New Haven, CT 065

Author Info: (1) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA. (2) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (3) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (4) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (5) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA. (6) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (7) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (8) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (9) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (10) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (11) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (12) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA. (13) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. (14) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA. (15) Gennao Bio, Hopewell, NJ 08525, USA. (16) Gennao Bio, Hopewell, NJ 08525, USA. (17) Gennao Bio, Hopewell, NJ 08525, USA. (18) Department of Medicine (Medical Oncology), Yale University School of Medicine, New Haven, CT 06520, USA. Department of Urology, Akita University School of Medicine, Akita, 010-8543, Japan. (19) Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA. (20) Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA. Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA. (21) Gennao Bio, Hopewell, NJ 08525, USA. (22) Gennao Bio, Hopewell, NJ 08525, USA. (23) Gennao Bio, Hopewell, NJ 08525, USA. (24) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA. (25) Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA. (26) Department of Medicine (Medical Oncology), Yale University School of Medicine, New Haven, CT 06520, USA. (27) Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA. Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA. Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA. (28) Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA. (29) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. Department of Medicine (Medical Oncology), Yale University School of Medicine, New Haven, CT 06520, USA. Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA. (30) Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA. Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA.

IL-12 mRNA-LNP promotes dermal resident memory CD4+ T cell development

Spotlight 

Zabala-Peñafiel, Gonzalez-Lombana et al. showed that mice vaccinated s.c. with LNPs containing mRNA encoding leishmanial PEPCK antigen generated specific dLN and splenic Th1 and T follicular helper cells, but few dermal resident memory T cells (dTrm). These mice did not mount delayed-type hypersensitivity (DTH, which requires dTrm in non-inflamed skin) or protective responses to L. major intradermal infection. Delivering IL-12 mRNA-LNPs with PEPCK vaccines boosted levels of specific Th1 cells with skin-homing (selectin+) and memory markers in LN, increased dTrm cells in inflamed and non-inflamed skin, and induced DTH and protective responses to Leishmania challenge.

Contributed by Paula Hochman

Zabala-Peñafiel, Gonzalez-Lombana et al. showed that mice vaccinated s.c. with LNPs containing mRNA encoding leishmanial PEPCK antigen generated specific dLN and splenic Th1 and T follicular helper cells, but few dermal resident memory T cells (dTrm). These mice did not mount delayed-type hypersensitivity (DTH, which requires dTrm in non-inflamed skin) or protective responses to L. major intradermal infection. Delivering IL-12 mRNA-LNPs with PEPCK vaccines boosted levels of specific Th1 cells with skin-homing (selectin+) and memory markers in LN, increased dTrm cells in inflamed and non-inflamed skin, and induced DTH and protective responses to Leishmania challenge.

Contributed by Paula Hochman

ABSTRACT: Dermal resident memory CD4(+) T cells (dTrm) provide protection against vector-borne infections. However, the factors that promote their development remain unclear. We tested if an mRNA vaccine, encoding a protective leishmanial antigen, induced dTrm cells. The mRNA vaccine induced robust systemic T-cell responses, but few Trm cells were found in the skin. Since IL-12 promotes Th1 responses, we tested whether IL-12 mRNA combined with the mRNA vaccine could enhance dTrm cell development. This combination significantly expanded Leishmania-specific Th1 cells expressing skin-homing molecules and memory T cell markers in the draining lymph node. Additionally, higher numbers of dTrm cells were maintained in the skin, and mice exhibited functional immunity indicated by a delayed hypersensitivity response and protection upon challenge with Leishmania. These findings highlight IL-12 as a key driver of CD4(+) dTrm development, enabling their global seeding across the skin, and underscore the potential of IL-12-enhanced mRNA vaccines to generate durable immunity against cutaneous leishmaniasis and other skin-targeted infections.

Author Info: (1) Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA. (2) Department of Pathobiology, School of Veterinary Medicine, Uni

Author Info: (1) Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA. (2) Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA. (3) Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA. (4) Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA. (5) Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada. (6) Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA. (7) Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA. (8) Acuitas Therapeutics, Vancouver, BC, Canada. (9) Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada. (10) Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA. Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA. (11) Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA. (12) Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA. pscott@upenn.edu.

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.