Journal Articles

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

Spotlight 

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

Contributed by Morgan Janes

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

Contributed by Morgan Janes

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

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

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

Intratumoral delivery of 4-1BBL boosts IL-12-triggered anti-glioma immunity Spotlight 

To overcome the immunosuppressive TME in glioblastoma, Lunavat and Nieland et al. identified the cell types involved in tumor control following i.t. IL-12 therapy in mice. Single cell RNAseq, targeted cell enrichment (CD45+, CD11b+), and depletion experiments demonstrated that among lymphocytes CD8+ were key to response to IL-12 and that a CCR7+ DC subset was also activated. Both cell types expressed 4-1BB and were further activatable by 4-1BBL delivered either by the implanted tumor cells transduced with Tnsf9 (encoding 4-1BBL) or by an intracerebrally administered AAV encoding 4-1BBL and actively expressed in GFAP+ astrocytes.

Contributed by Ed Fritsch

To overcome the immunosuppressive TME in glioblastoma, Lunavat and Nieland et al. identified the cell types involved in tumor control following i.t. IL-12 therapy in mice. Single cell RNAseq, targeted cell enrichment (CD45+, CD11b+), and depletion experiments demonstrated that among lymphocytes CD8+ were key to response to IL-12 and that a CCR7+ DC subset was also activated. Both cell types expressed 4-1BB and were further activatable by 4-1BBL delivered either by the implanted tumor cells transduced with Tnsf9 (encoding 4-1BBL) or by an intracerebrally administered AAV encoding 4-1BBL and actively expressed in GFAP+ astrocytes.

Contributed by Ed Fritsch

ABSTRACT: The standard of care in high-grade gliomas has remained unchanged in the past 20 years. Efforts to replicate effective immunotherapies in non-cranial tumors have led to only modest therapeutical improvements for patients with glioma. Here, we demonstrate that intratumoral (i.t.) administration of recombinant interleukin-12 (rIL-12) promotes local cytotoxic CD8(POS) T cell accumulation and conversion into an effector-like state, resulting in a dose-dependent survival benefit in preclinical glioblastoma (GB) mouse models. This tumor-reactive CD8 T cell response is further supported by intratumoral rIL-12-sensing dendritic cells (DCs) and is accompanied by the co-stimulatory receptor 4-1BB expression in both cell types. Given that DCs and CD8(POS) T cells are functionally suppressed in the tumor microenvironments (TME) of de novo and recurrent glioma patients, we tested whether anti-tumor response at the rIL-12-inflamed tumor site could be enhanced with 4-1BBL, the ligand of 4-1BB. 4-1BBL was delivered using an adeno-associated virus (AAV) vector targeting GFAP-expressing cells and resulted in prolonged survival of rIL-1 2-treated GB-bearing mice. This study establishes that tumor antigen (Ag)-specific CD8 T cell activity can be augmented by incorporating an AAV-vector-mediated gene therapy approach, effectively enhancing anti-GB immunity in the TME.

Author Info: (1) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; Department of Biomedicine, University o

Author Info: (1) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; Department of Biomedicine, University of Bergen, 5019 Bergen, Norway. (2) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; Department of Neurosurgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands. (3) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (4) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (5) Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Institute of Medical Engineering and Sciences and Department of Chemistry, Massachusetts Institute Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institutes of Technology, Cambridge, MA 02139, USA; Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada. (6) Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center at Harvard Medical School, Boston, MA 02215, USA. (7) Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center at Harvard Medical School, Boston, MA 02215, USA; Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. (8) Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (9) Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (10) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (11) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (12) Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA. (13) Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA. (14) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA. (15) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; Department of Neurosurgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands. (16) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (17) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (18) Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Brain Tumor Immunology and Immunotherapy Program, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA. (19) Harvard-MIT Health Sciences and Technology, Cambridge, MA 02139, USA. (20) Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Brain Tumor Immunology and Immunotherapy Program, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA. (21) Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA. (22) Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (23) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. (24) Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA. Electronic address: kbreyne@mgh.harvard.edu.

Transcriptional regulator SATB1 limits CD8+ T cell population expansion and effector differentiation in chronic infection and cancer

Spotlight 

Heyden et al. show that chromatin organizer SATB1 is enriched in TPEX cells and limits exhausted CD8+ T cell expansion and effector differentiation in chronic infection and cancer, while maintaining functionality. SATB1 downregulation is required for the differentiation into TEX cells with high TOX and GZMB expression. SATB1 regulated effector differentiation and central memory formation of CD8+ T cells in acute and chronic infection through direct DNA binding, independent of chromatin accessibility. KO in P14(CMV gp33) T cells increased TCF1+ TPEX and TEX cells accumulation in tumors, enhanced response to ICB, and improved survival in vivo.

Contributed by Shishir Pant

Heyden et al. show that chromatin organizer SATB1 is enriched in TPEX cells and limits exhausted CD8+ T cell expansion and effector differentiation in chronic infection and cancer, while maintaining functionality. SATB1 downregulation is required for the differentiation into TEX cells with high TOX and GZMB expression. SATB1 regulated effector differentiation and central memory formation of CD8+ T cells in acute and chronic infection through direct DNA binding, independent of chromatin accessibility. KO in P14(CMV gp33) T cells increased TCF1+ TPEX and TEX cells accumulation in tumors, enhanced response to ICB, and improved survival in vivo.

Contributed by Shishir Pant

ABSTRACT: CD8(+) T cells are major mediators of antiviral and antitumor immunity. During persistent antigen stimulation as in chronic infection and cancer, however, they differentiate into exhausted T cells that display impaired functionality. Precursors of exhausted T (T(PEX)) cells exhibit stem-like properties, including high proliferative, self-renewal and developmental potential, and are responsible for long-term CD8(+) T cell responses against persistent antigens. Here we identify the chromatin organizer and transcriptional regulator SATB1 as a major regulator of exhausted CD8(+) T cell differentiation. SATB1 was specifically expressed in T(PEX) cells where it limited population expansion and effector differentiation while preserving functionality of CD8(+) T cells. SATB1 downregulation was required for T(PEX) cell-to-effector cell differentiation in chronic infection and contributed to coordinated effector and memory differentiation in acute viral infection. DNA binding of SATB1 regulated gene expression both dependent and independent of chromatin accessibility. Finally, SATB1 limited antitumor CD8(+) and chimeric antigen receptor T cell immunity. Overall, our results identify SATB1 as a central regulator of precursor fate and effector differentiation of CD8(+) T cells both in infection and in cancer.

Author Info: (1) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. Institute of Molecul

Author Info: (1) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany. (2) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (3) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (4) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (5) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (6) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. Immunogenomics and Neurodegeneration, Deutsches Zentrum fŸr Neurodegenerative Erkrankungen (DZNE), Bonn, Germany. (7) Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia. (8) Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia. (9) Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia. (10) Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia. (11) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (12) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (13) Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany. (14) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (15) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (16) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (17) Immunogenomics and Neurodegeneration, Deutsches Zentrum fŸr Neurodegenerative Erkrankungen (DZNE), Bonn, Germany. (18) Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany. (19) Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany. (20) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (21) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (22) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (23) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (24) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. (25) Department of Microbiology, The Biomedical Discovery Institute, Monash University, Melbourne, Victoria, Australia. (26) Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia. (27) Immunogenomics and Neurodegeneration, Deutsches Zentrum fŸr Neurodegenerative Erkrankungen (DZNE), Bonn, Germany. Systems Medicine, Deutsches Zentrum fŸr Neurodegenerative Erkrankungen (DZNE), Bonn, Germany. PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE, University of Bonn, and West German Genome Center, Bonn, Germany (DZNE), Bonn, Germany. (28) Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany. (29) Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia. axel.kallies@unimelb.edu.au. Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany. axel.kallies@unimelb.edu.au.

Microbiota-derived butyrate promotes a FOXO1-induced stemness program and preserves CD8+ T cell immunity against melanoma

Spotlight 

Bachem, Clarke and Kong et al. studied the role of microbiota-derived short chain fatty acids (SCFAs) in an orthotopic melanoma model, revealing a direct link between melanoma control, CD8+ T cell differentiation and SCFA synthetic pathways. Dietary fiber and the SCFA butyrate reduced melanoma progression, increased tumor specific stem-like CD127+CD8+ T cells in the tdLN and induced a FOXO1-driven stemness program. In melanoma patients, metagenomic modeling of fecal samples revealed that increased butyrate flux correlated positively with ICB outcomes, consistent with previously observed correlations with dietary fiber consumption and fecal butyrate levels.

Contributed by Katherine Turner

Bachem, Clarke and Kong et al. studied the role of microbiota-derived short chain fatty acids (SCFAs) in an orthotopic melanoma model, revealing a direct link between melanoma control, CD8+ T cell differentiation and SCFA synthetic pathways. Dietary fiber and the SCFA butyrate reduced melanoma progression, increased tumor specific stem-like CD127+CD8+ T cells in the tdLN and induced a FOXO1-driven stemness program. In melanoma patients, metagenomic modeling of fecal samples revealed that increased butyrate flux correlated positively with ICB outcomes, consistent with previously observed correlations with dietary fiber consumption and fecal butyrate levels.

Contributed by Katherine Turner

ABSTRACT: A range of microbiota species correlate with improved cancer outcomes in patients and confer protection in pre-clinical mouse models. Here, we examined how microbiota regulate CD8(+) T cell immunity against melanoma. Spontaneous control of cutaneous melanoma in mice correlated with metabolic pathways required for microbial synthesis of short-chain fatty acids (SCFAs) shared between several microbiota species. Diet-induced enforcement of SCFA production by the gut microbiota reduced melanoma progression and enriched tumor-specific stem-like CD127(+)CD8(+) T cells in the tumor-draining lymph node (tdLN). The SCFA butyrate induced a FOXO1-driven stemness program and directly promoted the differentiation of tumor-specific CD127(+)CD8(+) T cells in the tdLN. Metabolic flux modeling predicted enhanced microbial production of butyrate in melanoma patients with complete therapeutic responses to immune checkpoint blockade (ICB), and butyrate induced transcriptional features of ICB responsiveness in CD8(+) T cells. Our findings suggest a critical role for metabolite production shared across several microbiota species in the preservation of stem-like tumor-specific CD8(+) T cells.

Author Info: (1) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. Electronic add

Author Info: (1) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. Electronic address: abachem@unimelb.edu.au. (2) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (3) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; Centre for Pathogen Genomics Innovation Hub, Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (4) Computational Science Initiative, Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (5) Computational Science Initiative, Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (6) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (7) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (8) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, North Rhine-Westphalia 53127, Germany. (9) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (10) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (11) Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia. (12) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (13) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (14) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (15) Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia. (16) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (17) Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia; Melanoma Discovery Laboratory, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia; School of Medicine, University of Western Australia, Crawley, WA 6009, Australia. (18) Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia; Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia. (19) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; Melbourne Integrative Genomics, School of BioSciences, The University of Melbourne, Melbourne, VIC 3010, Australia. (20) Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia. (21) Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia. (22) School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia; Telethon Kids Institute, The University of Western Australia, Perth, WA 6009, Australia. (23) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; Centre for Pathogen Genomics Innovation Hub, Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (24) Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia; Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia. (25) Computational Science Initiative, Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (26) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia. (27) Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; Institute of Innate Immunity, University of Bonn, Bonn, North Rhine-Westphalia 53127, Germany. Electronic address: sbedoui@unimelb.edu.au.

Clonally expanded effector CD4+ cytotoxic T lymphocytes are associated with severe neurological adverse events after immune checkpoint inhibitor therapy

Spotlight 

Giguelay and Maschmeyer et al. detected only minor differences in the PBMC immune cell composition of 17 cancer patients receiving ICI therapy, 8 with and 9 without severe neurological immune-related adverse events (n-irAEs). The subset most enriched in those with ICI-induced n-irAEs was clonally-expanded, terminally-differentiated CD4+ CTLs with upregulated expression of genes associated with antigen activation, neuroinflammation and cell lysis. Of potential biomarkers previously linked to ICI-induced n-irAEs, the CXCL10 receptor CXCR3 was upregulated in PBMC, but the frequency of the most expanded CD8+ T cell subset, (TEM cells) was not.

Contributed by Paula Hochman

Giguelay and Maschmeyer et al. detected only minor differences in the PBMC immune cell composition of 17 cancer patients receiving ICI therapy, 8 with and 9 without severe neurological immune-related adverse events (n-irAEs). The subset most enriched in those with ICI-induced n-irAEs was clonally-expanded, terminally-differentiated CD4+ CTLs with upregulated expression of genes associated with antigen activation, neuroinflammation and cell lysis. Of potential biomarkers previously linked to ICI-induced n-irAEs, the CXCL10 receptor CXCR3 was upregulated in PBMC, but the frequency of the most expanded CD8+ T cell subset, (TEM cells) was not.

Contributed by Paula Hochman

Background: Immune checkpoint inhibitor (ICI) therapies present a pillar of modern cancer therapy but can cause neurological immune-related adverse events (n-irAEs), of which up to 35% are severe or even fatal. However, the detailed immunological mechanisms and risk factors underlying n-irAEs remain largely unknown. Here, we leveraged single-cell genomics to dissect immune cell type, state, and clonal heterogeneity associated with n-irAEs.

Methods:
We performed coupled single-cell RNA sequencing and T cell receptor (TCR) profiling on peripheral blood cells of 17 patients with cancer receiving ICI therapy, including 8 patients with acute neurotoxicity. This approach enabled integrated analyses of immune cell states and T cell clonality linked to ICI-induced n-irAEs.

Results:
We profiled 186 435 immune cells and conducted pseudotime analyses, revealing that patients with n-irAEs, compared with controls, present with clonally expanded CD4+ cytotoxic T lymphocytes (CD4+ CTLs) with an n-irAE-specific effector gene expression profile. These T cells predominantly belong to a select set of expanded clonal families and express genes linked to antigen-induced activation, cell lysis, and neuroinflammation. Moreover, they highly express CXCR3 (FC=2.03 compared with control CD4+ CTLs, with a false discovery rate=7.7×10⁻⁴), encoding the chemokine receptor of CXCL10, previously nominated as a biomarker for severe ICI therapy-induced n-irAEs with concomitant multiple organ system toxicity.

Conclusions:
Overall, our study highlights the expansion and activation of CD4+ CTLs in ICI-induced neurotoxicity, proposing these cells as potential targets for developing new biomarkers and therapeutic strategies to improve patient outcomes.

Author Info: (1) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Max-DelbrŸck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Insti

Author Info: (1) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Max-DelbrŸck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany. (2) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Max-DelbrŸck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany. Institute for Systemic Inflammation Research (ISEF), University of LŸbeck, LŸbeck, Germany. (3) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Department of Neurology with Experimental Neurology, CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. (4) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Department of Neurology with Experimental Neurology, CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. (5) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Department of Neurology with Experimental Neurology, CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Einstein Center for Neurosciences Berlin (ECN) at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. (6) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Max-DelbrŸck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany. (7) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Max-DelbrŸck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany. Department of Biology, Chemistry, Pharmacy, Freie UniversitŠt Berlin, Berlin, Germany. Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA. (8) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Department of Neurology with Experimental Neurology, CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. NeuroCure Cluster of Excellence, CharitŽ UniversitŠtsmedizin Berlin, Berlin, Germany. (9) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Department of Neurology with Experimental Neurology, CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. NeuroCure Cluster of Excellence, CharitŽ UniversitŠtsmedizin Berlin, Berlin, Germany. (10) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. Department of Neurology with Experimental Neurology, CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. NeuroCure Cluster of Excellence, CharitŽ UniversitŠtsmedizin Berlin, Berlin, Germany. CharitŽ UniversitŠtsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany. German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Berlin, Germany. German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany. German Center for Mental Health (DZPG), partner site Berlin, Berlin, Germany. (11) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany leif.ludwig@bih-charite.de samuel.knauss@charite.de. Department of Neurology with Experimental Neurology, CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany. (12) Berlin Institute of Health at CharitŽ - UniversitŠtsmedizin Berlin, Berlin, Germany leif.ludwig@bih-charite.de samuel.knauss@charite.de. Max-DelbrŸck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany.

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

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

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

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

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

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

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

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

Contributed by Shishir Pant

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

Contributed by Shishir Pant

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

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

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

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

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 Spotlight 

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 Spotlight 

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.

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.