Expanding the cytokine receptor alphabet reprograms T cells into diverse states
(1) Zhao Y (2) Ogishi M (3) Pal A (4) Su LL (5) Tao P (6) Jiang H (7) Rodriguez GE (8) Chen X (9) Sun Q (10) Rysavy LW (11) Limsuwannarot S (12) Waghray D (13) Kalbasi A (14) Garcia KC
Matrix-M adjuvant triggers inflammasome activation and enables antigen cross-presentation through induction of lysosomal membrane permeabilization
(1) Zarnegar B (2) Carow B (3) Eriksson J (4) Spennare E (5) hlund P (6) Akpinar E (7) Bringeland E (8) Osterman IL (9) Lundqvist L (10) Antti J (11) Handin N (12) Helgesson PH (13) Bankefors J (14) Lvgren Bengtsson K (15) Sellin ME (16) Palm AE (17) Stertman L (18) Lunderius Andersson C
(1) Zarnegar B (2) Carow B (3) Eriksson J (4) Spennare E (5) hlund P (6) Akpinar E (7) Bringeland E (8) Osterman IL (9) Lundqvist L (10) Antti J (11) Handin N (12) Helgesson PH (13) Bankefors J (14) Lvgren Bengtsson K (15) Sellin ME (16) Palm AE (17) Stertman L (18) Lunderius Andersson C
Author Info: (1) Novavax AB, Uppsala, Sweden. (2) Novavax AB, Uppsala, Sweden. bcarow@novavax.com. (3) Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
Author Info: (1) Novavax AB, Uppsala, Sweden. (2) Novavax AB, Uppsala, Sweden. bcarow@novavax.com. (3) Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden. (4) Novavax AB, Uppsala, Sweden. (5) Novavax AB, Uppsala, Sweden. (6) Novavax AB, Uppsala, Sweden. (7) Novavax AB, Uppsala, Sweden. (8) Novavax AB, Uppsala, Sweden. (9) Novavax AB, Uppsala, Sweden. (10) Novavax AB, Uppsala, Sweden. (11) Novavax AB, Uppsala, Sweden. (12) Novavax AB, Uppsala, Sweden. (13) Novavax AB, Uppsala, Sweden. (14) Novavax AB, Uppsala, Sweden. (15) Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden. Science for Life Laboratory, Uppsala, Sweden. (16) Novavax AB, Uppsala, Sweden. (17) Novavax AB, Uppsala, Sweden. (18) Novavax AB, Uppsala, Sweden.
Citation: NPJ Vaccines 2025 Aug 5 10:184 Epub08/05/2025
Link to PUBMED: http://www.ncbi.nlm.nih.gov/pubmed/40764493
Succinate preserves CD8+ T cell fitness to augment antitumor immunity
Spotlight(1) Ma K (2) Cheng H (3) Wang L (4) Xiao H (5) Liu F (6) Yang Y (7) Xiao Z (8) Tang K (9) Li S (10) Wang G (11) Ge M (12) Wang J (13) Liu X (14) Sun HX (15) Luo Z (16) Gu Z (17) Ho PC (18) Li G (19) Zhang L
Ma et al. demonstrated that succinate promoted CD8+ T cell mitochondrial fitness and enhanced antitumor immunity. Succinate accumulation in tumors enhanced tumor-reactive CD8+ T cell-mediated immune responses. Succinate exposure promoted mitochondrial fitness through BNIP3-mediated mitophagy, and preserved T cell stemness via epigenetic remodeling. Adoptive transfer of succinate-treated T cells, including CAR T cells, displayed long-term persistence, synergized with anti-PD-L1 therapy, and showed superior antitumor activity in a melanoma tumor model. High succinate signatures correlated with better outcomes in ICB-treated patients.
Contributed by Shishir Pant
(1) Ma K (2) Cheng H (3) Wang L (4) Xiao H (5) Liu F (6) Yang Y (7) Xiao Z (8) Tang K (9) Li S (10) Wang G (11) Ge M (12) Wang J (13) Liu X (14) Sun HX (15) Luo Z (16) Gu Z (17) Ho PC (18) Li G (19) Zhang L
Ma et al. demonstrated that succinate promoted CD8+ T cell mitochondrial fitness and enhanced antitumor immunity. Succinate accumulation in tumors enhanced tumor-reactive CD8+ T cell-mediated immune responses. Succinate exposure promoted mitochondrial fitness through BNIP3-mediated mitophagy, and preserved T cell stemness via epigenetic remodeling. Adoptive transfer of succinate-treated T cells, including CAR T cells, displayed long-term persistence, synergized with anti-PD-L1 therapy, and showed superior antitumor activity in a melanoma tumor model. High succinate signatures correlated with better outcomes in ICB-treated patients.
Contributed by Shishir Pant
ABSTRACT: Succinate, a tricarboxylic acid cycle intermediate, accumulates in tumors with succinate dehydrogenase (SDH) mutations. Although succinate is recognized for modulating CD8(+) T cell cytotoxicity, its impact on T cell differentiation remains poorly understood. Here, we reveal that succinate accumulation in tumors lacking the SDH subunit B (SDHB) enhanced tumor-reactive CD8(+) T cell-mediated immune responses. Sustained succinate exposure promoted CD8(+) T cell survival and facilitated the generation and maintenance of stem-like subpopulations. Mechanistically, succinate enhanced mitochondrial fitness through Bcl-2/adenovirus E1B 19 kDa-interacting protein 3 (BNIP3)-mediated mitophagy and also promoted stemness-associated gene expression via epigenetic modulation. Succinate-conditioned CD8(+) T cells displayed superior long-term persistence and tumor control capacity. Moreover, succinate enrichment signature correlates with favorable clinical outcomes in certain melanoma and gastric cancer patients receiving immune checkpoint blockade therapy. These findings reveal how succinate preserves T cell stemness and highlight the therapeutic potential of succinate supplementation for enhancing T cell immunotherapy efficacy.
Author Info: (1) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu
Author Info: (1) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. (2) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. (3) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. (4) Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China. (5) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. (6) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215123, China. (7) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. (8) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Institute of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu 215123, China. (9) Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China. (10) Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China. (11) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. (12) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. (13) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. (14) College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China. (15) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. (16) State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. Electronic address: gzm@ism.pumc.edu.cn. (17) Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland. Electronic address: ping-chih.ho@unil.ch. (18) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. Electronic address: lgd@ism.cams.cn. (19) National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China. Electronic address: zlj@ism.cams.cn.
Citation: Immunity 2025 Aug 9 Epub08/09/2025
Link to PUBMED: http://www.ncbi.nlm.nih.gov/pubmed/40816269
Hypoimmune CD19 CAR T cells evade allorejection in patients with cancer and autoimmune disease Spotlight
(1) Hu X (2) Beauchesne P (3) Wang C (4) Wong A (5) Deuse T (6) Schrepfer S
14 patients with B cell cancers and 1 with autoimmune disease were treated with allogeneic anti-CD19 CAR T cells edited for immune evasion (TRAC/B2M/CIITA KD + CD47 overexpression), following lymphodepletion. The drug product was heterogeneous; fully edited CAR T (40.9% of cells) did not induce an immune response on-study, but incomplete editing resulted in T/NK cell cytotoxicity against CAR T cells, and donor-specific antibodies (DSA) that mediated cytotoxicity. Durable B cell depletion was observed in 6 patients, and was associated with increasing CAR T dose and DSA absence. 3 non-responding patients had pre-existing DSA, suggesting a potential biomarker.
Contributed by Alex Najibi
(1) Hu X (2) Beauchesne P (3) Wang C (4) Wong A (5) Deuse T (6) Schrepfer S
14 patients with B cell cancers and 1 with autoimmune disease were treated with allogeneic anti-CD19 CAR T cells edited for immune evasion (TRAC/B2M/CIITA KD + CD47 overexpression), following lymphodepletion. The drug product was heterogeneous; fully edited CAR T (40.9% of cells) did not induce an immune response on-study, but incomplete editing resulted in T/NK cell cytotoxicity against CAR T cells, and donor-specific antibodies (DSA) that mediated cytotoxicity. Durable B cell depletion was observed in 6 patients, and was associated with increasing CAR T dose and DSA absence. 3 non-responding patients had pre-existing DSA, suggesting a potential biomarker.
Contributed by Alex Najibi
ABSTRACT: Off-the-shelf CAR T cells need to reliably escape allogeneic immune responses to become universal medicines. The primary T cell product SC291 was engineered with a CD19 CAR, T cell receptor alpha constant (TRAC) knockout, and the hypoimmune (HIP) edits of HLA depletion and CD47 overexpression. Here, we report exploratory immune analyses from the ARDENT (NCT05878184) and GLEAM (NCT06294236) trials with HIP-edited CD19 CAR T cells. Although there was an alloimmune response against HLA-replete subpopulations of SC291, we observed no de novo immune response against fully edited HIP CAR T cells in all patients, irrespective of the dose or the patient's disease. The lack of antibodies against the HLA-replete CAR T cells was identified as a marker for deep tissue CD19 cell depletion, and all patients without such antibodies for 60 days showed concomitant B cell depletion in peripheral blood. The immune data presented support the reliability of the HIP concept to evade allorejection.
Author Info: (1) Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA, USA. (2) Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA, USA. (3) Sana Biotechnology Inc., 1 T
Author Info: (1) Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA, USA. (2) Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA, USA. (3) Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA, USA. (4) Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA, USA. (5) Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA, USA. (6) Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA, USA. Electronic address: sonja.schrepfer@sana.com.
Citation: Cell Stem Cell 2025 Aug 5 Epub08/05/2025
Link to PUBMED: http://www.ncbi.nlm.nih.gov/pubmed/40812299
Overcoming ovarian cancer resistance and evasion to CAR-T cell therapy by harnessing allogeneic CAR-NKT cells Spotlight
(1) Li YR (2) Li Z (3) Zhu Y (4) Li M (5) Chen Y (6) Lee D (7) Ochoa CJ (8) Singh T (9) DiBernardo G (10) Guo W (11) Li S (12) Zhou Y (13) Yu Y (14) Kramer A (15) Wilson M (16) Zhou K (17) Dunn ZS (18) Fang Y (19) Yu J (20) Kim YJ (21) Huang J (22) Shen X (23) Wang YC (24) Hon R (25) Basso T (26) Chen E (27) Zhu E (28) Lin S (29) Neal A (30) Moatamed NA (31) Oh SS (32) Sadeghi S (33) Pan C (34) Wang A (35) Lusis AJ (36) Zhou XJ (37) Zhou JJ (38) Pellegrini M (39) Wang P (40) Memarzadeh S (41) Yang L
Li et al. identified broad expression of NKR ligands on ovarian tumor cells, motivating the application of allogeneic, IL-15+ CAR NKT cells, which were effective in CAR antigen+/- xenograft models, and exhibited strong tumor homing and retention. CAR NKT cells sustained an effector/memory phenotype in vivo, and induced minimal changes in tumor cells, while CAR T cells became exhausted and led to immune-evasive alterations and antigen loss. CAR NKT cells also uniquely modified the TME through deletion of CD1d+ TAMs/MDSCs. Unlike CAR T cells, CAR NKT cells led to minimal CRS and did not induce GvHD, while also evading allorejection.
Contributed by Morgan Janes
(1) Li YR (2) Li Z (3) Zhu Y (4) Li M (5) Chen Y (6) Lee D (7) Ochoa CJ (8) Singh T (9) DiBernardo G (10) Guo W (11) Li S (12) Zhou Y (13) Yu Y (14) Kramer A (15) Wilson M (16) Zhou K (17) Dunn ZS (18) Fang Y (19) Yu J (20) Kim YJ (21) Huang J (22) Shen X (23) Wang YC (24) Hon R (25) Basso T (26) Chen E (27) Zhu E (28) Lin S (29) Neal A (30) Moatamed NA (31) Oh SS (32) Sadeghi S (33) Pan C (34) Wang A (35) Lusis AJ (36) Zhou XJ (37) Zhou JJ (38) Pellegrini M (39) Wang P (40) Memarzadeh S (41) Yang L
Li et al. identified broad expression of NKR ligands on ovarian tumor cells, motivating the application of allogeneic, IL-15+ CAR NKT cells, which were effective in CAR antigen+/- xenograft models, and exhibited strong tumor homing and retention. CAR NKT cells sustained an effector/memory phenotype in vivo, and induced minimal changes in tumor cells, while CAR T cells became exhausted and led to immune-evasive alterations and antigen loss. CAR NKT cells also uniquely modified the TME through deletion of CD1d+ TAMs/MDSCs. Unlike CAR T cells, CAR NKT cells led to minimal CRS and did not induce GvHD, while also evading allorejection.
Contributed by Morgan Janes
Author Info: (1) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los A
Author Info: (1) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (2) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (3) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (4) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (5) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (6) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (7) Department of Obstetrics and Gynecology, UCLA, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA. (8) Department of Obstetrics and Gynecology, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA. (9) Department of Obstetrics and Gynecology, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA. (10) Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA 90095, USA. (11) Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA. (12) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (13) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (14) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (15) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (16) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (17) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California (USC), Los Angeles, CA 90089, USA. (18) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (19) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (20) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (21) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (22) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (23) Division of Cardiology, Department of Medicine, UCLA, Los Angeles, CA 90095, USA. (24) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (25) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (26) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (27) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA. (28) Department of Biomedical Engineering, University of California, Davis (UCD), Davis, CA 95616, USA; Department of Surgery, School of Medicine, UCD, Sacramento, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA. (29) Department of Obstetrics and Gynecology, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA. (30) Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA. (31) Section of Interventional Pulmonology, Keck School of Medicine, USC, Los Angeles, CA 90089, USA. (32) Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA. (33) Division of Cardiology, Department of Medicine, UCLA, Los Angeles, CA 90095, USA. (34) Department of Biomedical Engineering, University of California, Davis (UCD), Davis, CA 95616, USA; Department of Surgery, School of Medicine, UCD, Sacramento, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA. (35) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Division of Cardiology, Department of Medicine, UCLA, Los Angeles, CA 90095, USA; Human Genetics, UCLA, Los Angeles, CA 90095, USA. (36) Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA. (37) Department of Biostatistics, Fielding School of Public Health, UCLA, Los Angeles, CA 90095, USA. (38) Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences-The Collaboratory, UCLA, Los Angeles, CA 90095, USA; Department of Molecular Cell and Developmental Biology, UCLA, Los Angeles, CA 90095, USA. (39) Mork Family Department of Chemical Engineering and Materials Science, University of Southern California (USC), Los Angeles, CA 90089, USA. (40) Department of Obstetrics and Gynecology, UCLA, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; The VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA; Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA. Electronic address: smemarzadeh@mednet.ucla.edu. (41) Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, UCLA, Los Angeles, CA 90095, USA; Goodman-Luskin Microbiome Center, UCLA, Los Angeles, CA 90095, USA. Electronic address: liliyang@ucla.edu.
Citation: Med 2025 Aug 5 100804 Epub08/05/2025
Link to PUBMED: http://www.ncbi.nlm.nih.gov/pubmed/40803321
Fc-optimized CD40 agonistic antibody elicits tertiary lymphoid structure formation and systemic antitumor immunity in metastatic cancer Featured
(1) Osorio JC (2) Knorr DA (3) Weitzenfeld P (4) Blanchard L (5) Yao N (6) Baez M (7) Sevilla C (8) DiLillo M (9) Rahman J (10) Sharma VP (11) Bromberg J (12) Postow MA (13) Ariyan C (14) Robson ME (15) Ravetch JV
In a phase 1 study, 12 patients with metastatic cancer were treated with intratumoral 2141-V11 – an Fc-engineered CD40 agonistic antibody with enhanced binding to the inhibitory receptor FcγRIIB. Treatment was safe and well tolerated. Six patients experienced tumor reduction, including two complete responses – one in melanoma and one in breast cancer. 2141-V11 induced regression in injected and non-injected lesions, demonstrating systemic and durable antitumor immunity. Complete responses were associated with systemic CD8+ T cell activation and the presence of tertiary lymphoid structures. Further mechanistic studies in mice agreed with these results.
(1) Osorio JC (2) Knorr DA (3) Weitzenfeld P (4) Blanchard L (5) Yao N (6) Baez M (7) Sevilla C (8) DiLillo M (9) Rahman J (10) Sharma VP (11) Bromberg J (12) Postow MA (13) Ariyan C (14) Robson ME (15) Ravetch JV
In a phase 1 study, 12 patients with metastatic cancer were treated with intratumoral 2141-V11 – an Fc-engineered CD40 agonistic antibody with enhanced binding to the inhibitory receptor FcγRIIB. Treatment was safe and well tolerated. Six patients experienced tumor reduction, including two complete responses – one in melanoma and one in breast cancer. 2141-V11 induced regression in injected and non-injected lesions, demonstrating systemic and durable antitumor immunity. Complete responses were associated with systemic CD8+ T cell activation and the presence of tertiary lymphoid structures. Further mechanistic studies in mice agreed with these results.
ABSTRACT: CD40 agonism enhances antitumor immunity but is limited by systemic toxicity and poor efficacy. Here, we present a phase 1 study (NCT04059588) of intratumoral (i.t.) 2141-V11, an Fc-engineered anti-CD40 agonistic antibody with enhanced binding to the inhibitory receptor Fc_RIIB. Among 12 metastatic cancer patients, 2141-V11 was well tolerated without dose-limiting toxicities. Six patients experienced tumor reduction, including two complete responses in melanoma and breast cancer. 2141-V11 induced regression in injected and non-injected lesions, correlating with systemic CD8(+) T cell activation and mature tertiary lymphoid structures (TLSs) in complete responders. In CD40/Fc_Rs humanized mice bearing orthotopic tumors, i.t. 2141-V11 promoted de novo TLS formation, facilitating i.t. CD8(+) T cell effector responses independent of lymph node priming. The resulting local immune responses by 2141-V11 mediated abscopal antitumor effects and sustained immune memory. These findings demonstrate that i.t. 2141-V11 is safe and promotes immune-privileged tumor microenvironments that promote systemic and durable antitumor immunity.
Author Info: (1) Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 1
Author Info: (1) Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA. Electronic address: osorioj@mskcc.org. (2) Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA. (3) Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA. (4) Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA. (5) Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (6) Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA. (7) Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA. (8) Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA. (9) Bio-Imaging Resource Center, Rockefeller University, New York, NY 10065, USA. (10) Bio-Imaging Resource Center, Rockefeller University, New York, NY 10065, USA. (11) Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (12) Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (13) Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (14) Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. (15) Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA. Electronic address: ravetch@rockefeller.edu.
Citation: Cancer Cell 2025 Aug 1 Epub08/01/2025
Link to PUBMED: http://www.ncbi.nlm.nih.gov/pubmed/40816292
Lung metastasis and recurrence is mitigated by CAR macrophages, in-situ-generated from mRNA delivered by small extracellular vesicles
(1) Xiao Y (2) Zhu T (3) Chen Z (4) Huang X
(1) Xiao Y (2) Zhu T (3) Chen Z (4) Huang X
Author Info: (1) Center for Infection and Immunity, Guangdong Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
Author Info: (1) Center for Infection and Immunity, Guangdong Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China. (2) Center for Infection and Immunity, Guangdong Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China. (3) Kingcell Regenerative Medicine (Guangdong) Co., Zhuhai, Guangdong, China. (4) Center for Infection and Immunity, Guangdong Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China. huangxi6@mail.sysu.edu.cn.
Citation: Nat Commun 2025 Aug 4 16:7166 Epub08/04/2025
Link to PUBMED: http://www.ncbi.nlm.nih.gov/pubmed/40759657
Humoral determinants of checkpoint immunotherapy Spotlight
(1) Dai Y (2) Aizenbud L (3) Qin K (4) Austin M (5) Jaycox JR (6) Cunningham J (7) Wang EY (8) Zhang L (9) Fischer S (10) Carroll SM (11) van Aggelen H (12) Kluger Y (13) Herold KC (14) Furchtgott L (15) Kluger HM (16) Ring AM
Dai, Aizenbud, and Qin et al. profiled the reactivities of autoantibodies (AAbs) from healthy donors and ICB-treated patients. AAb reactivities were diverse and often rare, differed between patients and healthy donors, generally did not associate with irAEs, and were largely unaffected by ICB. AAbs against inhibitory molecules, inflammatory cytokines, and surface TAAs associated with ICB response, while AAbs against costimulatory molecules or BMP receptors associated with non-responders. Functional inhibition by AAbs was validated in vitro, and blockade of AAb targets associated with responders (e.g., IFN-I and TL1A) supported ICB efficacy in mouse models.
Contributed by Alex Najibi
(1) Dai Y (2) Aizenbud L (3) Qin K (4) Austin M (5) Jaycox JR (6) Cunningham J (7) Wang EY (8) Zhang L (9) Fischer S (10) Carroll SM (11) van Aggelen H (12) Kluger Y (13) Herold KC (14) Furchtgott L (15) Kluger HM (16) Ring AM
Dai, Aizenbud, and Qin et al. profiled the reactivities of autoantibodies (AAbs) from healthy donors and ICB-treated patients. AAb reactivities were diverse and often rare, differed between patients and healthy donors, generally did not associate with irAEs, and were largely unaffected by ICB. AAbs against inhibitory molecules, inflammatory cytokines, and surface TAAs associated with ICB response, while AAbs against costimulatory molecules or BMP receptors associated with non-responders. Functional inhibition by AAbs was validated in vitro, and blockade of AAb targets associated with responders (e.g., IFN-I and TL1A) supported ICB efficacy in mouse models.
Contributed by Alex Najibi
ABSTRACT: Although the role of cellular immunity in checkpoint immunotherapy (CPI) for cancer is well established(1,2), the effect of antibody-mediated humoral immunity is comparably underexplored. Here we used rapid extracellular antigen profiling(3) to map the autoantibody reactome within a cohort of 374 patients with cancer treated with CPIs and 131 healthy control participants for autoantibodies to 6,172 extracellular and secreted proteins (the 'exoproteome'). Globally, patients with cancer treated with CPIs had diverse autoreactivities that were elevated relative to control individuals but changed minimally with treatment. Autoantibody signatures in patients treated with CPI strikingly distinguished them from healthy individuals. Although associations of specific autoantibodies with immune-related adverse events were sparse, we detected numerous individual autoantibodies that were associated with greatly altered odds ratios for response to therapy. These included autoantibodies to immunomodulatory proteins, such as cytokines, growth factors and immunoreceptors, as well as tumour surface proteins. Functional evaluation of several autoantibody responses indicated that they neutralized the activity of their target proteins, which included type I interferons (IFN-I), IL-6, OSM, TL1A, and BMPR1A and BMPR2. Modelling the effects of autoantibodies to IFN-I and TL1A in preclinical mouse tumour models resulted in enhanced CPI efficacy, consistent with their effects in patients. In conclusion, these findings indicate that autoantibodies to the exoproteome modify CPI responses and highlight therapeutically actionable pathways that can be exploited to augment immunotherapy.
Author Info: (1) Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA. (2) Yale Cancer Center, New Haven, CT, USA. (3) Division of Translational Science and Therapeutics, Fr
Author Info: (1) Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA. (2) Yale Cancer Center, New Haven, CT, USA. (3) Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA, USA. (4) Yale Cancer Center, New Haven, CT, USA. (5) Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA. (6) Program of Applied Mathematics, Yale University, New Haven, CT, USA. (7) Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA. (8) Yale Cancer Center, New Haven, CT, USA. (9) Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA. (10) Seranova Bio, South San Francisco, CA, USA. (11) Seranova Bio, South San Francisco, CA, USA. (12) Program of Applied Mathematics, Yale University, New Haven, CT, USA. Department of Pathology, Yale University School of Medicine, New Haven, CT, USA. (13) Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA. Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA. (14) Seranova Bio, South San Francisco, CA, USA. leon@seranovabio.com. (15) Yale Cancer Center, New Haven, CT, USA. harriet.kluger@yale.edu. (16) Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA, USA. aaronring@fredhutch.org.
Citation: Nature 2025 Jul 23 Epub07/23/2025
Link to PUBMED: http://www.ncbi.nlm.nih.gov/pubmed/40702172
Lymph-node-derived stem-like but not tumor-tissue-resident CD8+ T cells fuel anticancer immunity
Featured(1) Wijesinghe SKM (2) Rausch L (3) Gabriel SS (4) Galletti G (5) De Luca M (6) Qin L (7) Wen L (8) Tsui C (9) Man K (10) Heyden L (11) Mason T (12) Newland LD (13) Kueh A (14) Liao Y (15) Chisanga D (16) Swatler J (17) Voulaz E (18) Marulli G (19) Errico V (20) Losurdo A (21) Rossi GR (22) Souza-Fonseca-Guimaraes F (23) Huntington ND (24) Gebhardt T (25) Utzschneider DT (26) Herold MJ (27) Shi W (28) Schroeder J (29) Lugli E (30) Kallies A
Wijesinghe, Rausch, et al. studied various T cell subsets in genetic murine tumor models. Intratumor tissue resident-like CD8+ T cells were found not to be required for tumor control or ICB efficacy and could not differentiate into effector cells due to TGFβ exposure. In the tumor-draining lymph nodes, a population of precursors of exhausted CD8+ T cells was detected that was MYB-dependent. This population was essential for antitumor responses, had stem-like properties, and generated CX3CR1+ effector cells that migrated to tumors in response to ICB.
(1) Wijesinghe SKM (2) Rausch L (3) Gabriel SS (4) Galletti G (5) De Luca M (6) Qin L (7) Wen L (8) Tsui C (9) Man K (10) Heyden L (11) Mason T (12) Newland LD (13) Kueh A (14) Liao Y (15) Chisanga D (16) Swatler J (17) Voulaz E (18) Marulli G (19) Errico V (20) Losurdo A (21) Rossi GR (22) Souza-Fonseca-Guimaraes F (23) Huntington ND (24) Gebhardt T (25) Utzschneider DT (26) Herold MJ (27) Shi W (28) Schroeder J (29) Lugli E (30) Kallies A
Wijesinghe, Rausch, et al. studied various T cell subsets in genetic murine tumor models. Intratumor tissue resident-like CD8+ T cells were found not to be required for tumor control or ICB efficacy and could not differentiate into effector cells due to TGFβ exposure. In the tumor-draining lymph nodes, a population of precursors of exhausted CD8+ T cells was detected that was MYB-dependent. This population was essential for antitumor responses, had stem-like properties, and generated CX3CR1+ effector cells that migrated to tumors in response to ICB.
ABSTRACT: CD8(+) T cell-mediated tumor control and efficacy of immune checkpoint blockade (ICB) are associated with both precursors of exhausted T (T(PEX)) cells and tissue-resident memory T cells. Their relationships and relative contribution to tumor control, however, are insufficiently understood. Using single-cell RNA sequencing and genetic mouse models, we systematically dissected the heterogeneity and function of cytotoxic T cells in tumors and tumor-draining lymph nodes (tdLNs). We found that intratumoral TCF1(+) T(PEX) cells and their progeny acquired a tissue-residency program that limits their contribution to tumor control and ICB response. By contrast, MYB-dependent stem-like T(PEX) cells residing in tdLNs sustained CD8(+) T cell infiltration into tumors and mediated ICB response. The cytokine TGF_ was the central factor that enforced residency of intratumoral CD8(+) T cells and limited the abundance of stem-like T(PEX) cells in tdLNs, thereby restraining tumor control. A similar network of TGF_-constrained intratumoral and extratumoral CD8(+) T cells with precursor and residency characteristics was found in human cancer.
Author Info: (1) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (2) Department of
Author Info: (1) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (2) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (3) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (4) IRCCS Humanitas Research Hospital, Rozzano, Italy. School of Biological Sciences, Department of Molecular Biology, University of California, San Diego, San Diego, CA, USA. (5) IRCCS Humanitas Research Hospital, Rozzano, Italy. (6) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (7) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (8) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (9) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (10) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (11) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (12) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany. (13) The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia. Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia. School of Cancer Medicine, La Trobe University, Heidelberg, Victoria, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia. (14) Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia. School of Cancer Medicine, La Trobe University, Heidelberg, Victoria, Australia. Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia. (15) Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia. School of Cancer Medicine, La Trobe University, Heidelberg, Victoria, Australia. (16) IRCCS Humanitas Research Hospital, Rozzano, Italy. (17) IRCCS Humanitas Research Hospital, Rozzano, Italy. Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy. (18) IRCCS Humanitas Research Hospital, Rozzano, Italy. Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy. (19) IRCCS Humanitas Research Hospital, Rozzano, Italy. (20) IRCCS Humanitas Research Hospital, Rozzano, Italy. (21) Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland, Australia. (22) Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Queensland, Australia. (23) Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia. oNKo-Innate, Melbourne, Victoria, Australia. (24) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (25) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (26) The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia. Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia. School of Cancer Medicine, La Trobe University, Heidelberg, Victoria, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia. (27) Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia. School of Cancer Medicine, La Trobe University, Heidelberg, Victoria, Australia. Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia. (28) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. (29) IRCCS Humanitas Research Hospital, Rozzano, Italy. (30) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Queensland, Australia. axel.kallies@unimelb.edu.au. Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn, Bonn, Germany. axel.kallies@unimelb.edu.au.
Citation: Nat Immunol 2025 Aug 26:1367-1383 Epub07/29/2025
Link to PUBMED: http://www.ncbi.nlm.nih.gov/pubmed/40730900
Tags:
The tumor-sentinel lymph node immunomigratome reveals CCR7⁺ dendritic cells drive response to sequenced immunoradiotherapy
Spotlight(1) Saddawi-Konefka R (2) Msari RA (3) Tang S (4) Philips C (5) Sadat S (6) Clubb LM (7) Luna S (8) Fassardi S (9) Jones R (10) Pietryga IF (11) Faraji F (12) Schokrpur S (13) Yung BS (14) Allevato MM (15) Decker KE (16) Nasamran CA (17) Chilin-Fuentes D (18) Rosenthal SB (19) Jensen SM (20) Fox BA (21) Bell RB (22) Gutkind JS (23) Sharabi A (24) Califano JA
Using orthotopic 4MOSC murine models, Saddawi-Konefka et al. showed that the preferential migration of immune cells to the tumor-draining sentinel lymph nodes (SLNs) creates a unique immunologic niche with distinct cellular composition and transcriptional profiles. Sequencing tumor-directed lymphatic-sparing immunomodulatory radiation therapy prior to anti-PD-1 therapy enhanced local immunosurveillance, antigen presentation, and the efficacy of anti-PD-1 therapy, leading to a high rate of durable cures. MMP9-dependent entry of CCR7+ dendritic cells into the SLN was critical for the efficacy of tumor-directed immunoradiotherapy.
Contributed by Shishir Pant
(1) Saddawi-Konefka R (2) Msari RA (3) Tang S (4) Philips C (5) Sadat S (6) Clubb LM (7) Luna S (8) Fassardi S (9) Jones R (10) Pietryga IF (11) Faraji F (12) Schokrpur S (13) Yung BS (14) Allevato MM (15) Decker KE (16) Nasamran CA (17) Chilin-Fuentes D (18) Rosenthal SB (19) Jensen SM (20) Fox BA (21) Bell RB (22) Gutkind JS (23) Sharabi A (24) Califano JA
Using orthotopic 4MOSC murine models, Saddawi-Konefka et al. showed that the preferential migration of immune cells to the tumor-draining sentinel lymph nodes (SLNs) creates a unique immunologic niche with distinct cellular composition and transcriptional profiles. Sequencing tumor-directed lymphatic-sparing immunomodulatory radiation therapy prior to anti-PD-1 therapy enhanced local immunosurveillance, antigen presentation, and the efficacy of anti-PD-1 therapy, leading to a high rate of durable cures. MMP9-dependent entry of CCR7+ dendritic cells into the SLN was critical for the efficacy of tumor-directed immunoradiotherapy.
Contributed by Shishir Pant
ABSTRACT: Surgical ablation or broad radiation of tumor-draining lymph nodes can eliminate the primary tumor response to immunotherapy, highlighting the crucial role of these nodes in mediating the primary tumor response. Here, we show that immunoradiotherapy efficacy is dependent on treatment sequence and migration of modulated dendritic cells from tumor to sentinel lymph nodes. Using a tamoxifen-inducible reporter paired with CITE-sequencing in a murine model of oral cancer, we comprehensively characterize tumor immune cellular migration through lymphatic channels to sentinel lymph nodes at single-cell resolution, revealing a unique immunologic niche defined by distinct cellular phenotypic and transcriptional profiles. Through a structured approach of sequential immunomodulatory radiotherapy and checkpoint inhibition, we show that sequenced, lymphatic-sparing, tumor-directed radiotherapy followed by PD-1 inhibition achieves complete and durable tumor responses. Mechanistically, this treatment approach enhances migration of activated CCR7+ dendritic cell surveillance across the tumor-sentinel lymph node axis, revealing a shift from their canonical role in promoting tolerance to driving antitumor immunity. Overall, this work supports rationally sequencing immune-sensitizing, lymphatic-preserving, tumor-directed radiotherapy followed by immune checkpoint inhibition to optimize tumor response to immunoradiotherapy by driving activated dendritic cells to draining sentinel lymph nodes.
Author Info: (1) Department of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA, USA. rsaddawi@health.ucsd.edu. Moores Cancer Center, UC San Diego, La Jolla,
Author Info: (1) Department of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA, USA. rsaddawi@health.ucsd.edu. Moores Cancer Center, UC San Diego, La Jolla, CA, USA. rsaddawi@health.ucsd.edu. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. rsaddawi@health.ucsd.edu. (2) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. (3) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. (4) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. (5) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. (6) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Department of Pharmacology, UC San Diego, La Jolla, CA, USA. (7) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. (8) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. (9) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Department of Radiation Medicine and Applied Sciences, UC San Diego School of Medicine, San Diego, CA, USA. (10) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Department of Radiation Medicine and Applied Sciences, UC San Diego School of Medicine, San Diego, CA, USA. (11) Department of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA, USA. Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. (12) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Department of Medicine, Division of Hematology-Oncology, UC Davis, Sacramento, CA, USA. (13) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Department of Pharmacology, UC San Diego, La Jolla, CA, USA. (14) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Department of Pharmacology, UC San Diego, La Jolla, CA, USA. (15) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. (16) Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, CA, USA. (17) Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, CA, USA. (18) Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, CA, USA. (19) Earle A Chiles Research Institute, Robert W Franz Cancer Research Center, Providence Portland Medical Center, Portland, OR, USA. Department of Molecular Microbiology and Immunology, Oregon Health Science University, Portland, OR, USA. (20) Earle A Chiles Research Institute, Robert W Franz Cancer Research Center, Providence Portland Medical Center, Portland, OR, USA. Department of Molecular Microbiology and Immunology, Oregon Health Science University, Portland, OR, USA. (21) Earle A Chiles Research Institute, Robert W Franz Cancer Research Center, Providence Portland Medical Center, Portland, OR, USA. Department of Molecular Microbiology and Immunology, Oregon Health Science University, Portland, OR, USA. (22) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Department of Pharmacology, UC San Diego, La Jolla, CA, USA. (23) Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. Department of Radiation Medicine and Applied Sciences, UC San Diego School of Medicine, San Diego, CA, USA. (24) Department of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA, USA. jcalifano@health.ucsd.edu. Moores Cancer Center, UC San Diego, La Jolla, CA, USA. jcalifano@health.ucsd.edu. Gleiberman Head and Neck Cancer Center, Moores Cancer Center, UC San Diego, La Jolla, CA, USA. jcalifano@health.ucsd.edu.
Citation: Nat Commun 2025 Jul 17 16:6578 Epub07/17/2025
Link to PUBMED: http://www.ncbi.nlm.nih.gov/pubmed/40675962