Journal Articles

Multi-platform biomarkers of response to an immune checkpoint inhibitor in the neoadjuvant I-SPY 2 trial for early-stage breast cancer

Spotlight 

Campbell et al. analyzed biomarkers in pretreatment biopsies from patients with breast cancer in the I-SPY 2 trial (TN and HR+HER2- subtypes) to investigate immunological correlates of response to neoadjuvant anti-PD-1 therapy in the TME. Using 3 multiplex platforms, 126 biomarkers were evaluated and 56 were significantly associated with response to pembrolizumab. Tumors with high TILs and close spatial proximity of PD-1+ T cells and PD-L1+ cells (immune and tumor cells) correlated with a good response to pembrolizumab, including the TN and HR+HER2- subtypes, although different patterns of response were seen with different receptor subtypes.

Contributed by Katherine Turner

Campbell et al. analyzed biomarkers in pretreatment biopsies from patients with breast cancer in the I-SPY 2 trial (TN and HR+HER2- subtypes) to investigate immunological correlates of response to neoadjuvant anti-PD-1 therapy in the TME. Using 3 multiplex platforms, 126 biomarkers were evaluated and 56 were significantly associated with response to pembrolizumab. Tumors with high TILs and close spatial proximity of PD-1+ T cells and PD-L1+ cells (immune and tumor cells) correlated with a good response to pembrolizumab, including the TN and HR+HER2- subtypes, although different patterns of response were seen with different receptor subtypes.

Contributed by Katherine Turner

ABSTRACT: Only a subset of patients with breast cancer responds to immune checkpoint blockade (ICB). To better understand the underlying mechanisms, we analyze pretreatment biopsies from patients in the I-SPY 2 trial who receive neoadjuvant ICB using multiple platforms to profile the tumor microenvironment. A variety of immune cell populations and markers of immune/cytokine signaling associate with pathologic complete response (pCR). Interestingly, these differ by breast cancer receptor subtype. Measures of the spatial distributions of immune cells within the tumor microenvironment, in particular colocalization or close spatial proximity of PD-1+ T cells with PD-L1+ cells (immune and tumor cells), are significantly associated with response in the overall cohort as well as the in the triple negative (TN) and HR+HER2− subtypes. Our findings indicate that biomarkers associated with immune cell signaling, immune cell densities, and spatial metrics are predictive of neoadjuvant ICB efficacy in breast cancer.

Author Info: (1) Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA (2) Department of Laboratory Medicine, University of California, San Francisco, San

Author Info: (1) Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA (2) Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA (3) Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA (4) Biospecimen Resource Program (BIOS), University of California, San Francisco, San Francisco, CA 94143, USA (5) Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA (6) Akoya Biosciences, Marlborough, MA 01752, USA (7) Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA 95616, USA (8) Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA 95817, USA (9) Gemini Group, Ann Arbor, MI 48107, USA (10) Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA (11) Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637, USA (12) Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA (13) Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA (14) Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA (15) Department of Radiology, University of California, San Francisco, San Francisco, CA 94143, USA (16) Yale School of Medicine, Yale University, New Haven, CT 06510, USA (17) Berry Consultants, LLC, Austin, TX 78746, USA

A subset of neutrophils activates anti-tumor immunity and inhibits non-small-cell lung cancer progression

Spotlight 

Using IL-8hu models of NSCLC, Tang et al. found that IL-8 controlled tumor growth, and CD74hiSiglec6lo neutrophils emerged in response to IL-8 signaling. In vivo, this subset exhibited antigen cross-presentation capacity and superior T cell stimulation. CD74 KO ablated tumor control in IL-8 mice, reduced neutrophil MHC/CD86, and impaired T cell activation. Conversely, CD74 agonism slowed tumor growth in IL-8 mice alone and with anti-PD-L1 or osimertinib. A putative analogous human subset (CD74hiCD63lo) could be induced by IL-8 in vitro, exhibited superior T cell stimulation, and was more abundant in the blood of responders to ICB in NSCLC.

Contributed by Morgan Janes

Using IL-8hu models of NSCLC, Tang et al. found that IL-8 controlled tumor growth, and CD74hiSiglec6lo neutrophils emerged in response to IL-8 signaling. In vivo, this subset exhibited antigen cross-presentation capacity and superior T cell stimulation. CD74 KO ablated tumor control in IL-8 mice, reduced neutrophil MHC/CD86, and impaired T cell activation. Conversely, CD74 agonism slowed tumor growth in IL-8 mice alone and with anti-PD-L1 or osimertinib. A putative analogous human subset (CD74hiCD63lo) could be induced by IL-8 in vitro, exhibited superior T cell stimulation, and was more abundant in the blood of responders to ICB in NSCLC.

Contributed by Morgan Janes

ABSTRACT: Neutrophils in the tumor microenvironment (TME) are heterogeneous populations associated with cancer prognosis and immunotherapy. However, the plasticity and function of heterogeneous neutrophils in the TME of non-small-cell lung cancer (NSCLC) remain unclear. Here, we show that neutrophils produce high levels of interleukin (IL)-8, which induce the differentiation of CD74highSiglecFlow neutrophils and suppress the generation of CD74lowSiglecFhigh neutrophils in the TME of IL-8-humanized NSCLC mice. The CD74highSiglecFlow neutrophils boost anti-tumor T cell responses via antigen cross-presentation. Deleting CD74 in IL-8-humanized neutrophils impairs T cell activation and exacerbates NSCLC progression, whereas a CD74 agonist enhances T cell activation and the efficacy of anti-programmed cell death 1 (PD-1) or osimertinib therapies. Additionally, the CD74highCD63low neutrophils in the TME and peripheral blood of advanced NSCLC patients phenocopy the CD74highSiglecFlow neutrophils in the TME of NSCLC mice and correlate well with the responsiveness to anti-PD-1 plus chemotherapies. These findings demonstrate an IL-8-CD74high neutrophil axis that promotes anti-tumor immunity in NSCLC.

Author Info: (1) Department of Gastrointestinal Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan Universit

Author Info: (1) Department of Gastrointestinal Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China (2) Department of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China (3) Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China (4) Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (5) Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China (6) TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China

Probiotic neoantigen delivery vectors for precision cancer immunotherapy

Featured  

Redenti and Im et al. developed a novel system to deliver neoantigens using an engineered probiotic E. coli Nissle 1917, modified to enhance neoantigen expression, cytosolic delivery, and safety features, while minimizing immunosuppression in tumors. Delivered either i.t. or i.v., EcNcΔlonompT/LLO+ acted as an effective neoantigen vaccine, enhancing the accumulation and functionality of APCs, CD4+ T cells, and CD8+ T cells, and promoting effective and specific antitumor immunity that cleared and prevented tumors and extended survival in various mouse models.

Redenti and Im et al. developed a novel system to deliver neoantigens using an engineered probiotic E. coli Nissle 1917, modified to enhance neoantigen expression, cytosolic delivery, and safety features, while minimizing immunosuppression in tumors. Delivered either i.t. or i.v., EcNcΔlonompT/LLO+ acted as an effective neoantigen vaccine, enhancing the accumulation and functionality of APCs, CD4+ T cells, and CD8+ T cells, and promoting effective and specific antitumor immunity that cleared and prevented tumors and extended survival in various mouse models.

ABSTRACT: Microbial systems have been synthetically engineered to deploy therapeutic payloads in vivo1,2. With emerging evidence that bacteria naturally home in on tumours3,4 and modulate antitumour immunity5,6, one promising application is the development of bacterial vectors as precision cancer vaccines2,7. Here we engineered probiotic Escherichia coli Nissle 1917 as an antitumour vaccination platform optimized for enhanced production and cytosolic delivery of neoepitope-containing peptide arrays, with increased susceptibility to blood clearance and phagocytosis. These features enhance both safety and immunogenicity, achieving a system that drives potent and specific T cell-mediated anticancer immunity that effectively controls or eliminates tumour growth and extends survival in advanced murine primary and metastatic solid tumours. We demonstrate that the elicited antitumour immune response involves recruitment and activation of dendritic cells, extensive priming and activation of neoantigen-specific CD4+ and CD8+ T cells, broader activation of both T and natural killer cells, and a reduction of tumour-infiltrating immunosuppressive myeloid and regulatory T and B cell populations. Taken together, this work leverages the advantages of living medicines to deliver arrays of tumour-specific neoantigen-derived epitopes within the optimal context to induce specific, effective and durable systemic antitumour immunity.

Author Info: (1) Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA.(2) Department of Biomedical Engineering, Columbia

Author Info: (1) Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA.(2) Department of Biomedical Engineering, Columbia University, New York, NY, USA. (3) Department of Biomedical Engineering, Columbia University, New York, NY, USA. td2506@columbia.edu. (4) Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA. td2506@columbia.edu. (5) Data Science Institute, Columbia University, New York, NY, USA. td2506@columbia.edu. (6) Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA. na2697@cumc.columbia.edu. (7) Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA. na2697@cumc.columbia.edu. #Contributed equally.

Spatial dynamics of CD39+CD8+ exhausted T cell reveal tertiary lymphoid structures-mediated response to PD-1 blockade in esophageal cancer

Spotlight 

Tanoue et al. showed that in nivolumab-treated patients with ESCC, intratumoral CD39+PD-1+ CD8+ T cells were heterogeneous, comprising a precursor stem-like exhausted TCF1+(CD39+ Tpex) subpopulation localized to the stroma and concentrated adjacent to tertiary lymphoid structures, which differentiated and migrated to localize within the tumor parenchyma as TCF-1CD39+PD-1+ differentiated (more cytotoxic) Tex cells. ICB response was positively correlated with CD39+ Tpex cell abundance in tumors, but not with PD-L1 expression scores. Circulating proliferative CD39+ Tpex cell levels increased in responders following ICB therapy.

Contributed by Paula Hochman

Tanoue et al. showed that in nivolumab-treated patients with ESCC, intratumoral CD39+PD-1+ CD8+ T cells were heterogeneous, comprising a precursor stem-like exhausted TCF1+(CD39+ Tpex) subpopulation localized to the stroma and concentrated adjacent to tertiary lymphoid structures, which differentiated and migrated to localize within the tumor parenchyma as TCF-1CD39+PD-1+ differentiated (more cytotoxic) Tex cells. ICB response was positively correlated with CD39+ Tpex cell abundance in tumors, but not with PD-L1 expression scores. Circulating proliferative CD39+ Tpex cell levels increased in responders following ICB therapy.

Contributed by Paula Hochman

ABSTRACT: Despite the success of immune checkpoint blockade (ICB) therapy for esophageal squamous cell cancer, the key immune cell populations that affect ICB efficacy remain unclear. Here, imaging mass cytometry of tumor tissues from ICB-treated patients identifies a distinct cell population of CD39(+)PD-1(+)CD8(+) T cells, specifically the TCF1(+) subset, precursor exhausted T (CD39(+) Tpex) cells, which positively correlate with ICB benefit. CD39(+) Tpex cells are predominantly in the stroma, while differentiated CD39(+) exhausted T cells are abundantly and proximally within the parenchyma. Notably, CD39(+) Tpex cells are concentrated within and around tertiary lymphoid structure (TLS). Accordingly, tumors harboring TLSs have more of these cells in tumor areas than tumors lacking TLSs, suggesting Tpex cell recruitment from TLSs to tumors. In addition, circulating CD39(+) Tpex cells are also increased in responders following ICB therapy. Our findings show that this unique subpopulation of CD39(+)PD-1(+)CD8(+) T cells is crucial for ICB benefit, and suggest a key role in TLS-mediated immune responses against tumors.

Author Info: (1) Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (2) Department of Medicine and Biosystemic Science, Grad

Author Info: (1) Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (2) Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (3) Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (4) Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (5) Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (6) Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (7) Department of Hematology/Oncology, Japan Community Healthcare Organization Kyushu Hospital, Fukuoka, Japan. (8) Department of Imaging Science Program, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA. (9) Department of Medical Oncology, NHO National Hospital Organization Kyushu Medical Center, Fukuoka, Japan. (10) Department of Medical Oncology, Hamanomachi Hospital, Fukuoka, Japan. (11) Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (12) Department of Medical Oncology, Kitakyushu Municipal Medical Center, Fukuoka, Japan. (13) Department of Medical Oncology, Fukuoka Wajiro Hospital, Fukuoka, Japan. (14) Department of Medical Oncology, St Mary's Hospital, Kurume, Japan. (15) Department of Medical Oncology, Hamanomachi Hospital, Fukuoka, Japan. (16) Department of Gastrointestinal and Medical Oncology, National Kyushu Cancer Center, Fukuoka, Japan. (17) Department of Medical Oncology, Sasebo Kyosai Hospital, Nagasaki, Japan. (18) Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (19) Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (20) Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. Department of Pathology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan. (21) Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (22) Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. (23) Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. baba.eishi.889@m.kyushu-u.ac.jp.

P-stalk ribosomes act as master regulators of cytokine-mediated processes Spotlight 

Investigating how cytokine exposure rewires tumor cells, Dopler et al. found that activating cytokines like IFNγ, TNFα, IFNα/β, IL-1β, IL-6, and IL-17A, promoted the incorporation of P-stalk into ribosomes. Within cells, P-stalk ribosomes (PSRs) supported the preferential translation of mRNAs, particularly those encoding antigen processing and presentation machinery and other cytokine-regulated processes, enabling an “alert” state and increased susceptibility to immune detection and clearance. Loss of PSRs inhibited CD8+ T cell recognition and killing, and TGFβ was found to inhibit PSRs. P-stalk incorporation appeared to be transcriptionally regulated, while P-stalk inhibition was regulated by phosphorylation.

Contributed by Lauren Hitchings

Investigating how cytokine exposure rewires tumor cells, Dopler et al. found that activating cytokines like IFNγ, TNFα, IFNα/β, IL-1β, IL-6, and IL-17A, promoted the incorporation of P-stalk into ribosomes. Within cells, P-stalk ribosomes (PSRs) supported the preferential translation of mRNAs, particularly those encoding antigen processing and presentation machinery and other cytokine-regulated processes, enabling an “alert” state and increased susceptibility to immune detection and clearance. Loss of PSRs inhibited CD8+ T cell recognition and killing, and TGFβ was found to inhibit PSRs. P-stalk incorporation appeared to be transcriptionally regulated, while P-stalk inhibition was regulated by phosphorylation.

Contributed by Lauren Hitchings

ABSTRACT: Inflammatory cytokines are pivotal to immune responses. Upon cytokine exposure, cells enter an "alert state" that enhances their visibility to the immune system. Here, we identified an alert-state subpopulation of ribosomes defined by the presence of the P-stalk. We show that P-stalk ribosomes (PSRs) are formed in response to cytokines linked to tumor immunity, and this is at least partially mediated by P-stalk phosphorylation. PSRs are involved in the preferential translation of mRNAs vital for the cytokine response via the more efficient translation of transmembrane domains of receptor molecules involved in cytokine-mediated processes. Importantly, loss of the PSR inhibits CD8+ T cell recognition and killing, and inhibitory cytokines like transforming growth factor β (TGF-β) hinder PSR formation, suggesting that the PSR is a central regulatory hub upon which multiple signals converge. Thus, the PSR is an essential mediator of the cellular rewiring that occurs following cytokine exposure via the translational regulation of this process.

Author Info: (1) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (2) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (3)

Author Info: (1) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (2) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (3) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (4) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (5) Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (6) Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (7) Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (8) Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (9) Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (10) Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands. (11) Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (12) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (13) Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (14) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (15) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. (16) Proteomics Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands. (17) Proteomics Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands. (18) Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands. (19) Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (20) Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA. (21) Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands. (22) Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands. Electronic address: w.faller@nki.nl.

Programmable bacteria synergize with PD-1 blockade to overcome cancer cell-intrinsic immune resistance mechanisms Spotlight 

Li et al. engineered E. Coli Nissle to produce IFNγ with a synchronized integrated lysing circuit (SLIC-IFNγ), where at high density, the bacteria lyse and release IFNγ in recurring cycles. Relative to controls, i.t. injection of SLIC-IFNγ increased intratumoral IFNγ, TNFα+ and tumor- specific T cells, and antitumor efficacy, and also controlled contralateral tumors. In B2M- or JAK-deficient tumor models, or when injected i.v., SLIC-IFNγ remained effective and was further improved alongside anti-PD-1. Efficacy of i.t. SLIC-IFNγ in primary B2M-/- tumors was dependent on NK cells, and in contralateral WT tumors, relied on systemic T cell responses.

Contributed by Alex Najibi

Li et al. engineered E. Coli Nissle to produce IFNγ with a synchronized integrated lysing circuit (SLIC-IFNγ), where at high density, the bacteria lyse and release IFNγ in recurring cycles. Relative to controls, i.t. injection of SLIC-IFNγ increased intratumoral IFNγ, TNFα+ and tumor- specific T cells, and antitumor efficacy, and also controlled contralateral tumors. In B2M- or JAK-deficient tumor models, or when injected i.v., SLIC-IFNγ remained effective and was further improved alongside anti-PD-1. Efficacy of i.t. SLIC-IFNγ in primary B2M-/- tumors was dependent on NK cells, and in contralateral WT tumors, relied on systemic T cell responses.

Contributed by Alex Najibi

ABSTRACT: Interferon-γ (IFN-γ) is a potent cytokine critical for response to immunotherapy, yet conventional methods to systemically deliver this cytokine have been hindered by severe dose-limiting toxicities. Here, we engineered a strain of probiotic bacteria that home to tumors and locally release IFN-γ. A single intratumoral injection of these IFN-γ-producing bacteria was sufficient to drive systemic tumor antigen-specific antitumor immunity, without observable toxicity. Although cancer cells use various resistance mechanisms to evade immune responses, bacteria-derived IFN-γ overcame primary resistance to programmed cell death 1 (PD-1) blockade via activation of cytotoxic Foxp3-CD4+ and CD8+ T cells. Moreover, by activating natural killer (NK) cells, bacteria-derived IFN-γ also overcame acquired resistance mechanisms to PD-1 blockade, specifically loss-of-function mutations in IFN-γ signaling and antigen presentation pathways. Collectively, these results demonstrate the promise of combining IFN-γ-producing bacteria with PD-1 blockade as a therapeutic strategy for overcoming immunotherapy-resistant, locally advanced, and metastatic disease.

Author Info: (1) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. (2) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. (3) De

Author Info: (1) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. (2) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. (3) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. (4) Department of Biomedical Engineering, Columbia University, New York, NY, USA. (5) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. (6) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. (7) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. (8) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. (9) Department of Biomedical Engineering, Columbia University, New York, NY, USA. Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA. Data Science Institute, Columbia University, New York, NY, USA. (10) Department of Microbiology and Immunology, Columbia University, New York, NY, USA. Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.

Multimodal targeting chimeras enable integrated immunotherapy leveraging tumor-immune microenvironment Spotlight 

Lin et al. developed a flexible triple orthogonal linker platform to create multimodal targeting chimeras (Multi-TACs) that spatially coordinate immune cells within the tumor microenvironment through customizable linkers. The EGFR-CD3-PDL1 Multi-TAC, designed to co-engage T cells and dendritic cells at the tumor site, activated antigen-specific T cells and demonstrated robust antitumor efficacy in vitro, in several humanized mouse models, and in patient-derived tumor models. Multi-TAC was successfully expanded to enable tailored co-engagement of T–NK and T–myeloid cells, advancing immune-driven tumor control.

Contributed by Shishir Pant

Lin et al. developed a flexible triple orthogonal linker platform to create multimodal targeting chimeras (Multi-TACs) that spatially coordinate immune cells within the tumor microenvironment through customizable linkers. The EGFR-CD3-PDL1 Multi-TAC, designed to co-engage T cells and dendritic cells at the tumor site, activated antigen-specific T cells and demonstrated robust antitumor efficacy in vitro, in several humanized mouse models, and in patient-derived tumor models. Multi-TAC was successfully expanded to enable tailored co-engagement of T–NK and T–myeloid cells, advancing immune-driven tumor control.

Contributed by Shishir Pant

ABSTRACT: Although immunotherapy has revolutionized cancer treatment, its efficacy is affected by multiple factors, particularly those derived from the complexity and heterogeneity of the tumor-immune microenvironment (TIME). Strategies that simultaneously and synergistically engage multiple immune cells in TIME remain highly desirable but challenging. Herein, we report a multimodal and programmable platform that enables the integration of multiple therapeutic modules into single agents for tumor-targeted co-engagement of multiple immune cells within TIME. We developed the triple orthogonal linker (T-Linker) technology to integrate various therapeutic small molecules and biomolecules as multimodal targeting chimeras (Multi-TACs). The EGFR-CD3-PDL1 Multi-TAC facilitated T-dendritic cell co-engagement to target solid tumors with excellent efficacy, as demonstrated in vitro, in several humanized mouse models and in patient-derived tumor models. Furthermore, Multi-TACs were constructed to coordinate T cells with other immune cell types. The highly modular and programmable feature of our Multi-TACs may find broad applications in immunotherapy and beyond.

Author Info: (1) Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry o

Author Info: (1) Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Shenzhen Bay Laboratory, Shenzhen 518055, China. (2) Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. (3) National Resource Center for Mutant Mice, MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, Medical School of Nanjing University, Nanjing 210061, China. (4) Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. (5) Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China. (6) Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China. (7) State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China. (8) Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China. (9) Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Medical Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, China. (10) Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China. (11) Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China. (12) Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China. (13) Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China. (14) Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China. (15) Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China. (16) Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China. (17) Shenzhen Bay Laboratory, Shenzhen 518055, China. (18) Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. (19) Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China. Electronic address: linjian@pku.edu.cn. (20) Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210061, China; National Resource Center for Mutant Mice, MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, Medical School of Nanjing University, Nanjing 210061, China. Electronic address: yanli@nju.edu.cn. (21) Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China. Electronic address: kangxz@cicams.ac.cn. (22) Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. Electronic address: jzxi@pku.edu.cn. (23) Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Shenzhen Bay Laboratory, Shenzhen 518055, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China. Electronic address: pengchen@pku.edu.cn.

There are no spotlights this week

Spotlight 

Due to our extensive coverage of the conference, we will not include any spotlights this week.

Due to our extensive coverage of the conference, we will not include any spotlights this week.

Author Info:

Author Info:

Insights into the formulation of lipid nanoparticles for the optimization of mRNA therapeutics

mRNA-based therapeutics increasingly demonstrate significant potential in treating various diseases, including infectious diseases, cancers, and genetic disorders. Effective delivery systems are crucial for advancing mRNA therapeutics. Lipid nanoparticles (LNPs) serve as an excellent carrier, widely validated for their safety and tolerability in commercially available mRNA vaccines. Standard LNPs typically consist of four components: ionizable lipids (ILs), helper lipids, cholesterol, and polyethylene glycol-lipids (PEG-lipids), with the structural design of ILs gradually becoming a focal point of research interest. The chemical structures and formulations of the other components also significantly affect the delivery efficiency, targeting specificity, and stability of LNPs. The complex formulations of LNPs may hinder the clinical transformation of mRNA therapeutics and have raised widespread concerns about their safety. This review aims to summarize the progress of LNPs-based mRNA therapeutics in clinical trials, focusing on adverse effects that occurred during these trials. It also discusses representative innovations in LNP components, highlighting challenges and potential ways in this research field. We firmly believe this review will promote further improvements and designs of LNP compositions to optimize mRNA therapeutics. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Biology-Inspired Nanomaterials > Lipid-Based Structures.

Author Info: (1) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (2) Department of Critical

Author Info: (1) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (2) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (3) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (4) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (5) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (6) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (7) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (8) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (9) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China. (10) Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.

Glutamine availability regulates cDC subsets in tissue

Proliferating tumor cells take up glutamine for anabolic processes engendering glutamine deficiency in the tumor microenvironment. How this might impact immune cells is not well understood. Using multiple mouse models of soft tissue sarcomas, glutamine antagonists, as well as genetic and pharmacological inhibition of glutamine utilization, we found that the number and frequency of conventional dendritic cells (cDC) is dependent on microenvironmental glutamine levels. cDCs comprise two distinct subsets - cDC1 and cDC2, with the former subset playing a critical role in antigen cross-presentation and tumor immunity. While both subsets show dependence on Glutamine, cDC1s are particularly sensitive. Notably, glutamine antagonism did not reduce the frequency of DC precursors but decreased proliferation and survival of cDC1s. Further studies suggest a role of the nutrient sensing mTOR signaling pathway in this process. Taken together, these findings uncover glutamine dependence of cDC1s that is coopted by tumors to escape immune responses. ONE SENTENCE SUMMARY: Type 1 conventional dendritic cells require glutamine to maintain their number in non-lymphoid tissue. SIGNIFICANCE: Immune evasion is a key hallmark of cancer; however, the underlying pathways are diverse, tumor-specific and not fully elucidated. Many tumor cells avidly import glutamine to support their anabolic needs, creating a glutamine-deficient tumor microenvironment (TME). Herein, using mouse models of soft tissue sarcomas, we show that glutamine depletion in TME leads to reduced type 1 conventional dendritic cells - a cell type that is critical for adaptive immune responses. This work is a paradigm for how tumor cell metabolism can regulate anti-tumor immune responses and will be foundational to future efforts targeting glutamine metabolism for cancer immunotherapy.

Author Info: (1) (2) (3) (4) (5) (6) (7) (8) (9) (10) (11)

Author Info: (1) (2) (3) (4) (5) (6) (7) (8) (9) (10) (11)

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.