Journal Articles

Collagen-binding IL-12-armoured STEAP1 CAR-T cells reduce toxicity and treat prostate cancer in mouse models

Sasaki et al. addressed the toxicity of IL-12 that has caused dose-limiting immune-related adverse events (irAEs). Compared to unmodified IL-12, fusing a collagen-binding domain to IL-12 (CBD–IL-12) improved the potency of CAR T cells targeting STEAP1 in both mouse and human prostate cancer models by (1) enhancing intratumoral IFNγ levels and IL-12 retention in the TME, and (2) reducing irAEs, such as hepatotoxicity and T cell infiltration into non-target organs. In an established mouse prostate tumor model, CBD–IL-12 armored CAR T cells combined with checkpoint inhibitors showed strong antitumor efficacy, and extended survival with immune memory.

Contributed by Katherine Turner

Sasaki et al. addressed the toxicity of IL-12 that has caused dose-limiting immune-related adverse events (irAEs). Compared to unmodified IL-12, fusing a collagen-binding domain to IL-12 (CBD–IL-12) improved the potency of CAR T cells targeting STEAP1 in both mouse and human prostate cancer models by (1) enhancing intratumoral IFNγ levels and IL-12 retention in the TME, and (2) reducing irAEs, such as hepatotoxicity and T cell infiltration into non-target organs. In an established mouse prostate tumor model, CBD–IL-12 armored CAR T cells combined with checkpoint inhibitors showed strong antitumor efficacy, and extended survival with immune memory.

Contributed by Katherine Turner

ABSTRACT: Immunosuppressive microenvironments, the lack of immune infiltration, and antigen heterogeneity pose challenges for chimaeric antigen receptor (CAR)-T cell therapies applied to solid tumours. Previously, CAR-T cells were armoured with immunostimulatory molecules, such as interleukin 12 (IL-12), to overcome this issue, but faced high toxicity. Here we show that collagen-binding domain-fused IL-12 (CBD-IL-12) secreted from CAR-T cells to target human six transmembrane epithelial antigen of prostate 1 (STEAP1) is retained within murine prostate tumours. This leads to high intratumoural interferon-_ levels, without hepatotoxicity and infiltration of T cells into non-target organs compared with unmodified IL-12. Both innate and adaptive immune compartments are activated and recognize diverse tumour antigens after CBD-IL-12-armoured CAR-T cell treatment. A combination of CBD-IL-12-armoured CAR-T cells and immune checkpoint inhibitors eradicated large tumours in an established prostate cancer mouse model. In addition, human CBD-IL-12-armoured CAR-T cells showed potent anti-tumour efficacy in a 22Rv1 xenograft while reducing circulating IL-12 levels compared with unmodified IL-12-armoured CAR-T cells. CBD fusion to potent payloads for CAR-T therapy may remove obstacles to their clinical translation towards elimination of solid tumours.

Author Info: (1) Department of Bioengineering, Imperial College London, London, UK. (2) Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. Division of Hematology/Oncology,

Author Info: (1) Department of Bioengineering, Imperial College London, London, UK. (2) Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA. (3) Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. (4) Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. (5) Department of Bioengineering, Imperial College London, London, UK. (6) Department of Bioengineering, Imperial College London, London, UK. (7) Department of Bioengineering, Imperial College London, London, UK. (8) Department of Bioengineering, Imperial College London, London, UK. (9) Department of Bioengineering, Imperial College London, London, UK. (10) Department of Microbiology, Immunology and Molecular Genetics, UCLA, Duarte, CA, USA. (11) Department of Bioengineering, Imperial College London, London, UK. (12) Department of Bioengineering, Imperial College London, London, UK. (13) Department of Bioengineering, Imperial College London, London, UK. (14) Department of Bioengineering, Imperial College London, London, UK. (15) Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA. (16) Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA. (17) Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA. Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA. (18) Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. Division of Medical Oncology, University of Washington, Seattle, WA, USA. (19) Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. jklee@mednet.ucla.edu. Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA. jklee@mednet.ucla.edu. Parker Institute for Cancer Immunotherapy at UCLA, Los Angeles, CA, USA. jklee@mednet.ucla.edu. (20) Department of Bioengineering, Imperial College London, London, UK. j.ishihara@imperial.ac.uk. Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba, Japan. j.ishihara@imperial.ac.uk.

Engineering T cells with a membrane-tethered version of SLP-76 overcomes antigen-low resistance to CAR T cell therapy

Compared to TCRs, CARs have poorer recruitment of proximal signaling molecules and antigen (Ag) sensitivity. To improve CAR T cell responses in low-Ag settings, Rotiroti et al. demonstrated that membrane-bound SLP-76 (MT-SLP-76), but not cytosolic SLP-76, improved CAR T cell cytokine responses to diverse Ag levels. In vivo, MT-SLP-76 improved CAR T cell expansion and efficacy in Ag-low models (CD22, CD19, BCMA) and maintained persistence in Ag-high models. MT-SLP-76 was mediated through ITK and PLCγ1, and enriched cytokine pathway expression. MT-SLP-76 also increased the potential for on-target, off-tumor toxicity, which could narrow the therapeutic window.

Contributed by Alex Najibi

Compared to TCRs, CARs have poorer recruitment of proximal signaling molecules and antigen (Ag) sensitivity. To improve CAR T cell responses in low-Ag settings, Rotiroti et al. demonstrated that membrane-bound SLP-76 (MT-SLP-76), but not cytosolic SLP-76, improved CAR T cell cytokine responses to diverse Ag levels. In vivo, MT-SLP-76 improved CAR T cell expansion and efficacy in Ag-low models (CD22, CD19, BCMA) and maintained persistence in Ag-high models. MT-SLP-76 was mediated through ITK and PLCγ1, and enriched cytokine pathway expression. MT-SLP-76 also increased the potential for on-target, off-tumor toxicity, which could narrow the therapeutic window.

Contributed by Alex Najibi

ABSTRACT: Chimeric antigen receptor (CAR) T cells can mediate durable complete responses in individuals with certain hematologic malignancies, but antigen downregulation is a common mechanism of resistance. Although the native T cell receptor can respond to very low levels of antigen, engineered CARs cannot, likely due to inefficient recruitment of downstream proximal signaling molecules. We developed a platform that endows CAR T cells with the ability to kill antigen-low cancer cells consisting of a membrane-tethered version of the cytosolic signaling adaptor molecule SLP-76 (MT-SLP-76). MT-SLP-76 can be expressed alongside any CAR to lower its activation threshold, overcoming antigen-low escape in multiple xenograft models. Mechanistically, MT-SLP-76 amplifies CAR signaling through recruitment of ITK and PLC_1. MT-SLP-76 was designed based on biologic principles to render CAR T cell therapies less susceptible to antigen downregulation and is poised for clinical development to overcome this common mechanism of resistance.

Author Info: (1) Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. (2) Department of Genetics, Stanford University, Stanford, CA, USA. Department of Medicine, Sta

Author Info: (1) Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. (2) Department of Genetics, Stanford University, Stanford, CA, USA. Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA. (3) Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. (4) Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA. (5) Proteas Health, Torrance, CA, USA. (6) Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. (7) Department of Pathology, Stanford University, Stanford, CA, USA. (8) Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. (9) Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. (10) Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. (11) Department of Radiation Oncology and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA. (12) Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. (13) Immunology Graduate Program, Stanford University School of Medicine, Stanford, CA, USA. Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA. (14) Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA. (15) Immuno-Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. (16) Immuno-Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA. (17) Department of Pathology, Stanford University, Stanford, CA, USA. Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA. (18) Department of Pediatrics, City of Hope National Medical Center, Duarte, CA, USA. Department of Immuno-oncology, City of Hope National Medical Center, Duarte, CA, USA. (19) Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. (20) Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. robbie_majzner@dfci.harvard.edu. Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA. robbie_majzner@dfci.harvard.edu. Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA. robbie_majzner@dfci.harvard.edu. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. robbie_majzner@dfci.harvard.edu.

Neoadjuvant immunotherapy in mismatch-repair-proficient colon cancers

Tan et al. reported clinical outcomes and translational analyses of patients with early-stage DNA mismatch repair-proficient colon cancer from the phase II NICHE trial. Out of 33 patients, 17 received nivolumab and ipilimumab alone and 16 received them in combination with celecoxib. The overall response rate was 26% with 6 MPR and 1 CR. Circulating tumor DNA (ctDNA) was negative in 5/6 responders after neoadjuvant treatment and prior to surgery, while 19/20 non-responders remained ctDNA-positive. Responders showed higher chromosomal genomic instability, increased TCF1 expression, and proliferation of CD8+ T cells, despite a low TMB in all tumors.

Contributed by Shishir Pant

Tan et al. reported clinical outcomes and translational analyses of patients with early-stage DNA mismatch repair-proficient colon cancer from the phase II NICHE trial. Out of 33 patients, 17 received nivolumab and ipilimumab alone and 16 received them in combination with celecoxib. The overall response rate was 26% with 6 MPR and 1 CR. Circulating tumor DNA (ctDNA) was negative in 5/6 responders after neoadjuvant treatment and prior to surgery, while 19/20 non-responders remained ctDNA-positive. Responders showed higher chromosomal genomic instability, increased TCF1 expression, and proliferation of CD8+ T cells, despite a low TMB in all tumors.

Contributed by Shishir Pant

ABSTRACT: Immune checkpoint blockade (ICB) has led to paradigm shifts in the treatment of various tumour types(1-4), yet limited efficacy has been observed in patients with metastatic mismatch-repair proficient (pMMR) colorectal cancer(5). Here we report clinical results and in-depth analysis of patients with early-stage pMMR colon cancer from the phase II NICHE study (ClinicalTrials.gov: NCT03026140). A total of 31 patients received neoadjuvant treatment of nivolumab plus ipilimumab followed by surgery. The response rate was 26% and included six patients with a major pathological response (²10% residual viable tumour). One patient with an ongoing clinical complete response did not undergo surgery. Circulating tumour DNA (ctDNA) was positive in 26/31 patients at baseline, and clearance was observed in 5/6 responders prior to surgery, while 19/20 non-responders remained ctDNA+. Responses were observed despite a low tumour mutational burden in all tumours, while chromosomal genomic instability scores were significantly higher in responders compared to non-responders. Furthermore, responding tumours had significantly higher baseline expression of proliferation signatures and TCF1, and imaging mass cytometry revealed a higher percentage of Ki-67(+) cancer and Ki-67(+) CD8(+) T cells in responders compared to non-responders. These results provide a comprehensive analysis of response to neoadjuvant ICB in early-stage pMMR colon cancers and identify potential biomarkers for patient selection.

Author Info: (1) Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (2) Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Am

Author Info: (1) Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (2) Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (3) Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (4) Department of Biometrics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (5) Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands. (6) Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands. (7) Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands. (8) Natera, Inc, Austin, TX, USA. (9) Natera, Inc, Austin, TX, USA. (10) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (11) Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands. (12) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (13) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Oncode Institute, Utrecht, the Netherlands. (14) Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands. (15) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Oncode Institute, Utrecht, the Netherlands. Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands. (16) Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands. Oncode Institute, Utrecht, the Netherlands. (17) Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. m.chalabi@nki.nl. Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. m.chalabi@nki.nl.

SARS-CoV-2 mRNA vaccines sensitize tumours to immune checkpoint blockade

Grippin, Marconi, Copling, Li, et al. discovered that COVID-19 mRNA vaccination within 100 days of ICB treatment initiation resulted in improved survival in patients with NSCLC and metastatic melanoma. In murine models, this effect was mechanistically found to relate to increased type I IFN signaling, which activated APCs, increased tumor antigen presentation, and increased tumor-reactive CD8+ T cell activation in lymphoid organs and tumors. In human tumors, tumoral PD-L1 expression increased and survival improved in those vaccinated within 100 days of ICB initiation, even in those with very low PD-L1 expression in NSCLC at baseline.

Grippin, Marconi, Copling, Li, et al. discovered that COVID-19 mRNA vaccination within 100 days of ICB treatment initiation resulted in improved survival in patients with NSCLC and metastatic melanoma. In murine models, this effect was mechanistically found to relate to increased type I IFN signaling, which activated APCs, increased tumor antigen presentation, and increased tumor-reactive CD8+ T cell activation in lymphoid organs and tumors. In human tumors, tumoral PD-L1 expression increased and survival improved in those vaccinated within 100 days of ICB initiation, even in those with very low PD-L1 expression in NSCLC at baseline.

ABSTRACT: Immune checkpoint inhibitors (ICIs) extend survival in many patients with cancer but are ineffective in patients without pre-existing immunity(1-9). Although personalized mRNA cancer vaccines sensitize tumours to ICIs by directing immune attacks against preselected antigens, personalized vaccines are limited by complex and time-intensive manufacturing processes(10-14). Here we show that mRNA vaccines targeting SARS-CoV-2 also sensitize tumours to ICIs. In preclinical models, SARS-CoV-2 mRNA vaccines led to a substantial increase in type I interferon, enabling innate immune cells to prime CD8(+) T cells that target tumour-associated antigens. Concomitant ICI treatment is required for maximal efficacy in immunologically cold tumours, which respond by increasing PD-L1 expression. Similar correlates of vaccination response are found in humans, including increases in type I interferon, myeloid-lymphoid activation in healthy volunteers and PD-L1 expression on tumours. Moreover, receipt of SARS-CoV-2 mRNA vaccines within 100_days of initiating ICI is associated with significantly improved median and three-year overall survival in multiple large retrospective cohorts. This benefit is similar among patients with immunologically cold tumours. Together, these results demonstrate that clinically available mRNA vaccines targeting non-tumour-related antigens are potent immune modulators capable of sensitizing tumours to ICIs.

Author Info: (1) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. agrippin@mdanderson.org. (2) Lillian S. W

Author Info: (1) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. agrippin@mdanderson.org. (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) McGovern Medical School, Houston, TX, USA. (4) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (5) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (6) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (7) Department of Enterprise Data Engineering and Analytics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (8) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (9) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (10) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (11) Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, 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) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, 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) McGovern Medical School, Houston, TX, USA. (17) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, 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. Department of Pediatrics, Division of Hematology-Oncology, University of Florida, Gainesville, FL, USA. (20) Department of Dermatology, Baylor College of Medicine, Houston, TX, USA. (21) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (22) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (23) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (24) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (25) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (26) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (27) Department of Pathology, Stanford University, Stanford, CA, USA. (28) McGovern Medical School, Houston, TX, USA. (29) McGovern Medical School, Houston, TX, USA. (30) Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (31) Scripps Health, San Diego, CA, USA. (32) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (33) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 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) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (36) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (37) University of North Carolina, Chapel Hill, NC, USA. (38) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (39) Department of Surgical Oncology, Division Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (40) Department of Surgical Oncology, Division Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (41) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (42) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (43) Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (44) Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (45) Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (46) Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (47) Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (48) Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (49) Department of Biostatistics, The University of Florida, Gainesville, FL, USA. (50) Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (51) Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (52) Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (53) Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (54) Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (55) Department of Surgical Oncology, Division Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (56) Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (57) Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (58) Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA. (59) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. (60) 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 Hematology-Oncology, University of Florida, Gainesville, FL, USA. elias.sayour@neurosurgery.ufl.edu. (61) Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. shlin@mdanderson.org.

A pan-immunotherapy signature to predict intratumoral CD8+ T cell expansions

Takahashi et al. established a multi-site murine tumor model system for temporal tracking of CD8+ T cell phenotypes and clonal dynamics, identifying a “precursor exhausted” PD-1+Ly108+ population as a driver of clonal expansion. DEG analysis revealed an “expansion signature” that correlated with future expansion in baseline samples from patients treated with ICB. Therapeutic responses and survival were associated with higher expansion scores in post-treatment, but not pre-treatment samples. In the murine model, LAG3 blockade uniquely enhanced the total expansion signature by selectively re-expanding previously contracted existing clones.

Contributed by Morgan Janes

Takahashi et al. established a multi-site murine tumor model system for temporal tracking of CD8+ T cell phenotypes and clonal dynamics, identifying a “precursor exhausted” PD-1+Ly108+ population as a driver of clonal expansion. DEG analysis revealed an “expansion signature” that correlated with future expansion in baseline samples from patients treated with ICB. Therapeutic responses and survival were associated with higher expansion scores in post-treatment, but not pre-treatment samples. In the murine model, LAG3 blockade uniquely enhanced the total expansion signature by selectively re-expanding previously contracted existing clones.

Contributed by Morgan Janes

ABSTRACT: Effective cancer immunotherapy relies on the clonal proliferation and expansion of CD8(+) T cells in the tumor. However, our insights into clonal expansions are limited, owing to an inability to track the same clones in tumors over time. Here, we develop a multi-site tumor mouse model system to track hundreds of expanding and contracting CD8(+) T cell clones over multiple timepoints in tumors of the same individual. Through coupling of clonal expansion dynamics and single-cell RNA/TCR-seq data, we identify a transcriptomic signature in PD-1(+)Ly108(+) precursor exhausted cells that strongly predicts rates of intratumoral clone expansion. The signature correlates with expansion in mice, both with and without immunotherapies, and in patients undergoing PD-1 blockade therapy. Expression of the signature during treatment corresponds with positive clinical outcomes. Downregulation of the signature precedes clone contraction-a phase in which clones contract but maintain revivable precursor exhausted cells in the tumor. LAG-3 blockade re-activates the expansion signature, re-expanding pre-existing clones, including previously contracted clones. These findings reveal how the study of clonal expansion dynamics provide a powerful 'pan-immunotherapy' signature for monitoring immunotherapies with implications for their future development.

Author Info: (1) Medical Research Council Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK. munetomo.takahashi@gmail.com. Department of Prev

Author Info: (1) Medical Research Council Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK. munetomo.takahashi@gmail.com. Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. munetomo.takahashi@gmail.com. (2) Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan. (3) Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan. Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA. (4) Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan. (5) Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan. (6) Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan. (7) Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK. (8) Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. (9) Medical Research Council Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK. (10) Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan. (11) Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan. ueha@rs.tus.ac.jp.

αTIGIT-IL2 achieves tumor regression by promoting tumor-infiltrating regulatory T cell fragility in mouse models

Wang et al. generated αTIGIT-IL2, an immunocytokine comprising IL-2 and an anti-TIGIT scFv fused to a human IgG1Fc. Within tumors, αTIGIT-IL2 (delivered i.p.) preferentially bound to Tregs (due to their high CD25 expression), reduced Treg inhibitory activity, and stimulated Treg production of IFNγ to promote a “hot” TME. The enhanced inflammatory environment of the TME recruited and activated neutrophils to be proinflammatory and act as APCs to boost CD8+ T cell antitumor activity in multiple “cold” murine tumor models. αTIGIT-IL2 and a blocking PD-1 Ab synergized to inhibit ICB-resistant TNBC tumor growth and induce immune memory.

Contributed by Paula Hochman

Wang et al. generated αTIGIT-IL2, an immunocytokine comprising IL-2 and an anti-TIGIT scFv fused to a human IgG1Fc. Within tumors, αTIGIT-IL2 (delivered i.p.) preferentially bound to Tregs (due to their high CD25 expression), reduced Treg inhibitory activity, and stimulated Treg production of IFNγ to promote a “hot” TME. The enhanced inflammatory environment of the TME recruited and activated neutrophils to be proinflammatory and act as APCs to boost CD8+ T cell antitumor activity in multiple “cold” murine tumor models. αTIGIT-IL2 and a blocking PD-1 Ab synergized to inhibit ICB-resistant TNBC tumor growth and induce immune memory.

Contributed by Paula Hochman

ABSTRACT: Administration of IL-2 may promote the suppressive function and proliferation of Treg cells that cause immune tolerance in patients with cancer, which causes low-dose IL-2 to fail in achieving an optimal anti-tumor effect. Here, we designed an immunocytokine by fusing IL-2 and an anti-TIGIT monoclonal antibody, named αTIGIT-IL2, that targets Treg cells and promotes their fragility in the tumor milieu. These fragile-like Treg cells show impaired suppressive function and high IFN-γ production, triggering an immune-reactive tumor microenvironment. Such inflammation leads to the recruitment and functional reprogramming of intratumoral neutrophils, improving cross-talk between neutrophils and CD8+ T cells and enhancing the antitumor ability of CD8+ T cells. Combination therapy with αTIGIT-IL2 and PD-1 blocker could eliminate triple-negative breast cancer (TNBC) tumors resistant to immune checkpoint blockade (ICB) therapy. These findings provide the basis for developing a new generation of immunocytokines that target Treg cells and promote their fragility in the tumor milieu, resulting in robust antitumor immunity.

Author Info: (1) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Scie

Author Info: (1) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. (2) MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. (3) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. (4) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. (5) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. (6) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. (7) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. (8) Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China. (9) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. Hefei TG ImmunoPharma Corporation Limited, Hefei, China. (10) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. mahongdi@ustc.edu.cn. (11) Hefei TG ImmunoPharma Corporation Limited, Hefei, China. haoyusun@ustc.edu.cn. Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China. haoyusun@ustc.edu.cn. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. haoyusun@ustc.edu.cn. Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China. haoyusun@ustc.edu.cn. (12) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. tzg@ustc.edu.cn. Hefei TG ImmunoPharma Corporation Limited, Hefei, China. tzg@ustc.edu.cn. Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China. tzg@ustc.edu.cn. (13) State Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. ustczxh@ustc.edu.cn. Hefei TG ImmunoPharma Corporation Limited, Hefei, China. ustczxh@ustc.edu.cn. Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China. ustczxh@ustc.edu.cn.

Spatial immune profiling defines a subset of human gliomas with functional tertiary lymphoid structures Spotlight 

Using spatial multiomics, Cakmak et al. profiled 642 adult-type diffuse gliomas and identified tertiary lymphoid structures (TLSs) in 15% of the tumors. TLS-positive gliomas showed decreased neural tumor signatures, and were associated with vascular and extracellular matrix remodeling. Three distinct TLS subtypes – T-low-TLS (low T cell abundance), B:T-TLS (B and T cells interaction), and PC-TLS (plasma cell-rich) – with different cellular compositions and immune functions were identified. B:T-TLS and PC-TLS showed lymphocyte clonal expansion, plasma cell differentiation, and DC–T cell interactions, and were associated with increased survival.

Contributed by Shishir Pant

Using spatial multiomics, Cakmak et al. profiled 642 adult-type diffuse gliomas and identified tertiary lymphoid structures (TLSs) in 15% of the tumors. TLS-positive gliomas showed decreased neural tumor signatures, and were associated with vascular and extracellular matrix remodeling. Three distinct TLS subtypes – T-low-TLS (low T cell abundance), B:T-TLS (B and T cells interaction), and PC-TLS (plasma cell-rich) – with different cellular compositions and immune functions were identified. B:T-TLS and PC-TLS showed lymphocyte clonal expansion, plasma cell differentiation, and DC–T cell interactions, and were associated with increased survival.

Contributed by Shishir Pant

ABSTRACT: Adult-type diffuse gliomas, the most common primary brain tumors, respond poorly to immune-based therapies and are considered immunologically "cold" tumors. Here, we examined the features and clinical relevance of glioma intratumoral tertiary lymphoid structures (TLSs) using spatial transcriptome and proteome profiling. In a cohort of 642 gliomas, TLSs were present in 15% of tumors and associated with a remodeled perivascular space and spatial redistribution of extracellular matrix components. Three distinct TLS subtypes could be defined based on differing cellular composition and immune activity. While all subtypes lacked classical germinal center architecture, certain TLSs exhibited features of dynamic immune functions, including clonal T and B cell expansion, generation of IgA_ and IgG_ plasma cells, and dendritic cell-T cell interactions. The presence of TLSs with active immune response features correlated with improved overall survival. Thus, a functional adaptive immune response is detectable in some gliomas, with implications for stratification and treatment.

Author Info: (1) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany. (2) Goethe Univers

Author Info: (1) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany. (2) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany. (3) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany; University Cancer Center (UCT), Frankfurt, Germany. (4) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany. (5) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany. (6) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany; University Cancer Center (UCT), Frankfurt, Germany. (7) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany; University Cancer Center (UCT), Frankfurt, Germany. (8) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany. (9) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany. (10) Frankfurt Cancer Institute (FCI), Frankfurt, Germany; University Cancer Center (UCT), Frankfurt, Germany; Goethe University, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt, Germany. (11) University Cancer Center (UCT), Frankfurt, Germany; Goethe University, University Hospital, Institute of Neuroradiology, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany. (12) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany; University Cancer Center (UCT), Frankfurt, Germany. (13) Frankfurt Cancer Institute (FCI), Frankfurt, Germany; University Cancer Center (UCT), Frankfurt, Germany; Goethe University, University Hospital, Institute of Neuroradiology, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany. (14) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany; University Cancer Center (UCT), Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany. (15) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany. (16) Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Frankfurt, Germany; University Cancer Center (UCT), Frankfurt, Germany. Electronic address: imkeller@rz.uni-frankfurt.de.

Targeting SALL4 with an HLA Class I-restricted TCR for cancer immunotherapy

Khelil et al. demonstrated that SALL4 is overexpressed in multiple tumor types, including primary colorectal cancers and paired liver metastasis, but is silenced in almost all adult tissues. The SALL4-derived S9V peptide was restricted by HLA-A2, and induced specific CD8+ T cell responses from the peripheral blood of patients with GI cancers. In vitro, SALL4-S9V-specific TCR-engineered CD8+ T cells were cytotoxic against SALL4-expressing tumor cells, but didn’t recognize hematopoietic stem cells (HSC). Adoptively transferred SALL4-S9V-specific TCR T cells suppressed tumor growth and improved survival in SALL4+ MDA-MB231 xenografts, without affecting HSCs.

Contributed by Shishir Pant

Khelil et al. demonstrated that SALL4 is overexpressed in multiple tumor types, including primary colorectal cancers and paired liver metastasis, but is silenced in almost all adult tissues. The SALL4-derived S9V peptide was restricted by HLA-A2, and induced specific CD8+ T cell responses from the peripheral blood of patients with GI cancers. In vitro, SALL4-S9V-specific TCR-engineered CD8+ T cells were cytotoxic against SALL4-expressing tumor cells, but didn’t recognize hematopoietic stem cells (HSC). Adoptively transferred SALL4-S9V-specific TCR T cells suppressed tumor growth and improved survival in SALL4+ MDA-MB231 xenografts, without affecting HSCs.

Contributed by Shishir Pant

ABSTRACT: Aberrant expression of the oncogene SALL4 is associated with stemness, more aggressive cancer phenotype, and reduced patient survival in various tumor types making SALL4 a potential target for cancer immunotherapy. We conducted a transcriptional analysis of SALL4 expression in colorectal cancer (CRC) tissues and demonstrated that SALL4 was overexpressed in primary tumor and paired liver metastasis. Then, we identified the SALL4-derived S9V peptide as a naturally processed peptide that induced specific CD8+ T-cell responses from the peripheral blood of gastrointestinal cancer patients whereas no responses were observed for the peripheral blood of healthy donors. Thereafter, we isolated a SALL4-specific T-cell receptor (TCR) that recognized this peptide in the most common HLA molecule in the Caucasian population, HLA-A2, and used this to develop TCR-engineered T cells. In vitro analysis showed that SALL4 TCR-redirected primary CD8+ T cells exhibited cytotoxic effects against SALL4-expressing tumor cells and produced effector cytokines. In vivo, SALL4-TCR T cells significantly reduced tumor growth and improved survival of tumor-bearing mice. Moreover, SALL4-TCR T cells displayed no toxicity against hematopoietic stem cells. Thus, we conclude that T cells engineered to express a SALL4-specific TCR have the potential to be effective as immunotherapy for solid cancers and pave the way for further clinical development.

Author Info: (1) Institut Gustave Roussy, Villejuif, France. (2) INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comte, Besançon, France, Besançon, Bourgogne-Franche-Comte, France. (3) Un

Author Info: (1) Institut Gustave Roussy, Villejuif, France. (2) INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comte, Besançon, France, Besançon, Bourgogne-Franche-Comte, France. (3) Université Bourgogne Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France, Besançon, France. (4) Université Bourgogne Franche-Comté, France. (5) Université Bourgogne Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France, Besançon, France. (6) Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France, Besançon, France. (7) Université Marie et Louis Pasteur, EFS, INSERM UMR1098 RIGHT, Besançon, F-25000, France, Besançon, Bourgogne-Franche-Comte, France. (8) Université Bourgogne Franche-Comté, France. (9) University of Lausanne, Lausanne, Switzerland. (10) Interactions Hte-Greffon-Tumeur & Ingénierie Cellulaire et Génique, Besançon, France. (11) Memorial Sloan Kettering Cancer Center, France. (12) Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France, Besançon, France. (13) University Hospital of Besançon, Besançon, France. (14) EFS-BFC, Besançon, France. (15) Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France, Besançon, Bourgogne-Franche-Comté, France. (16) Ludwig Institute for Cancer Research and Department of Oncology, University of Lausanne, Lausanne, Switzerland. (17) University Hospital of Lausanne, Lausanne, Switzerland. (18) University of Geneva, Geneva, Switzerland. (19) Université de Franche-Comté, CHU Besançon, EFS, INSERM, UMR RIGHT, F-25000 Besan̤on, France, Besançon, France. (20) INSERM CIC-1431, Clinical Investigation Center in Biotherapy, University Hospital of Besançon, F-25000 Besançon, France, France. (21) INSERM, UMR1098, Besançon cedex, France. (22) University of Franche-Comté, Besançon, France. (23) University Hospital of Besançon, France. (24) university hospital of Besançon, Besançon, France. (25)Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France, Besançon, France.

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

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

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

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

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

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

Contributed by Shishir Pant

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

Contributed by Shishir Pant

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

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

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

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.