Journal Articles

Survivin Dendritic Cell Vaccine Safely Induces Immune Responses and Is Associated with Durable Disease Control after Autologous Transplant in Patients with Myeloma

PURPOSE: We investigated whether a dendritic cell (DC) vaccine transduced with an adenoviral vector encoded with full-length survivin (Ad-S), with mutations neutralizing its antiapoptotic function, could safely generate an immune response and deepen clinical responses when administered before and after autologous stem cell transplant (ASCT) for multiple myeloma. PATIENTS AND METHODS: This phase I first-in-human trial (NCT02851056) evaluated the safety of DC:Ad-S in newly diagnosed multiple myeloma not having achieved complete response with induction, given 7 to 30 days prior to stem cell collection and 20 to 34 days after ASCT. Anti-survivin antibodies and CD4+ and CD8+ specific T cells were quantified. RESULTS: A total of 14 patients were treated and 13 included in the primary efficacy analysis. No serious adverse events were attributed to DC:Ad-S vaccine. Detectable anti-survivin antibodies increased from baseline in 9 of 13 (69%) patients, and 11 of 13 (85%) mounted either a cellular or humoral immune response to survivin. Seven patients had an improved clinical response at day +90, all of whom had mounted an immune response, and 6 of 7 patients remain event-free at a median follow-up of 4.2 years. Estimated progression-free survival at 4 years is 71% (95% confidence interval, 41-88). CONCLUSIONS: Two doses of DC:Ad-S, one given immediately before and another after ASCT, were feasible and safe. A high frequency of vaccine-specific immune responses was seen in combination with durable clinical outcomes, supporting ongoing investigation into the potential of this approach.

Author Info: (1) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (2) Department of Blood and Marrow Transplant and Cellular Immunoth

Author Info: (1) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (2) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (3) Department of Blood and Marrow Transplant and Cellular Immunotherapy, University of Virginia, Charlottesville, Virginia. (4) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (5) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (6) Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida. (7) Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida. (8) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (9) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (10) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (11) Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida. (12) Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida. (13) Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida. (14) Department of Immunology, Moffitt Cancer Center, Tampa, Florida. (15) Department of Pathology, Moffitt Cancer Center, Tampa, Florida. (16) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (17) Department of Medicine, Duke University, Durham, North Carolina. (18) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida. (19) Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.

Baseline interleukin-6 is a prognostic factor for patients with metastatic breast cancer treated with eribulin

PURPOSE: Eribulin is a unique anti-cancer drug which can improve overall survival (OS) of patients with metastatic breast cancer (MBC), probably by modulating the tumor immune microenvironment. The aim of this study was to investigate the clinical significance of serum levels of immune-related and inflammatory cytokines in patients treated with eribulin. Furthermore, we investigated the association between cytokines and immune cells, such as myeloid-derived suppressor cells (MDSCs) and cytotoxic and regulatory T cells, to explore how these cytokines might affect the immune microenvironment. METHODS: Sixty-eight patients with MBC treated with eribulin were recruited for this retrospective study. The relationship of cytokines, including interleukin (IL)-6, to progression-free survival and OS was examined. CD4(+)_and CD8(+)_lymphocyte, MDSCs and regulatory T cell levels were determined in the blood by flow cytometry analysis. RESULTS: In our cohort, patients with high IL-6 at baseline had shorter progression-free survival and OS compared with those with low IL-6 (p_=_0.0017 and p_=_0.0012, respectively). Univariable and multivariable analyses revealed that baseline IL-6 was an independent prognostic factor for OS (p_=_0.0058). Importantly, CD8(+)_lymphocytes were significantly lower and MDSCs were significantly higher in patients with high IL-6, compared to those with low IL-6. CONCLUSION: Baseline IL-6 is an important prognostic factor in patients with MBC treated with eribulin. Our results show that high IL-6 is associated with higher levels of MDSCs which suppress anti-tumor immunity, such as CD8(+)_cells. It appears that eribulin is not particularly effective in patients with high IL-6 due to a poor tumor immune microenvironment.

Author Info: (1) Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan. (2) Depart

Author Info: (1) Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan. (2) Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan. (3) Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan. (4) Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan. (5) Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan. ymiyoshi@hyo-med.ac.jp.

Cytotoxicity of WT1-reactive T cells against Wilms tumor: An implication for antigen-specific adoptive immunotherapy

INTRODUCTION: T cells that recognize WT1 peptides have been shown to efficiently eliminate WT1-expressing tumor cells. This study was designed to investigate the feasibility of isolating WT1-reactive T cells from peripheral blood mononuclear cells (PBMCs) from healthy donors and patients with Wilms tumor, and to assess the cytotoxicity mediated by these cells against Wilms tumor cells (WiTu cells). METHODS: WT1-reactive T cells were enriched and isolated by stimulating PBMCs with a WT1 peptide pool and interferon-_ capture-based immunomagnetic separation (IMS). Using the lactate dehydrogenase release assay, the in vitro cytotoxicity of the isolated cells and standard chemotherapy was evaluated on WiTu cells. RESULTS: Higher proportions of WT1-reactive T cells were isolated from patients with Wilms tumor compared to those isolated from HDs. WT1-reactive T cells produced > 50% specific lysis when co-cultured with WT1(+) WiTu cells at the highest effector-to-target (E:T) ratio in this study (i.e., 5:1), compared to <23% when co-cultured with WT1(-) WiTu cells at the same ratio. WT1-reactive T cells showed anti-tumoral activity in a dose-dependent manner and mediated significantly greater cytotoxicity than the non-WT1-reactive fraction of PBMCs on WT1(+) WiTu cells. The cytotoxicity of standard chemotherapy was significantly lower than that of WT1-reactive T cells when co-cultured with WT1(+) WiTu cells at E:T ratios of 2:1 and 5:1. CONCLUSION: WT1-reactive T cells can be effectively enriched from the PBMCs of patients with Wilms tumor. Ex vivo generated WT1-reactive T cells might be considered an adoptive immunotherapeutic option for WT1(+) Wilms tumors.

Author Info: (1) Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran. Cancer Control Foundation, Iran Universit

Author Info: (1) Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran. Cancer Control Foundation, Iran University of Medical Sciences, Tehran, Iran. Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran. (2) Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran. (3) Department of Pathology, Tehran University of Medical Sciences, Tehran, Iran. (4) Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran. (5) Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. (6) Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran. (7) Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran. Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. (8) Cancer Control Foundation, Iran University of Medical Sciences, Tehran, Iran. Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.

Current advances in cancer vaccines targeting NY-ESO-1 for solid cancer treatment

New York-esophageal cancer 1 (NY-ESO-1) belongs to the cancer testis antigen (CTA) family, and has been identified as one of the most immunogenic tumor-associated antigens (TAAs) among the family members. Given its ability to trigger spontaneous humoral and cellular immune response and restricted expression, NY-ESO-1 has emerged as one of the most promising targets for cancer immunotherapy. Cancer vaccines, an important element of cancer immunotherapy, function by presenting an exogenous source of TAA proteins, peptides, and antigenic epitopes to CD4(+) T cells via major histocompatibility complex class II (MHC-II) and to CD8(+) T cells via major histocompatibility complex class I (MHC-I). These mechanisms further enhance the immune response against TAAs mediated by cytotoxic T lymphocytes (CTLs) and helper T cells. NY-ESO-1-based cancer vaccines have a history of nearly two decades, starting from the first clinical trial conducted in 2003. The current cancer vaccines targeting NY-ESO-1 have various types, including Dendritic cells (DC)-based vaccines, peptide vaccines, protein vaccines, viral vaccines, bacterial vaccines, therapeutic whole-tumor cell vaccines, DNA vaccines and mRNA vaccines, which exhibit their respective benefits and obstacles in the development and application. Here, we summarized the current advances in cancer vaccines targeting NY-ESO-1 for solid cancer treatment, aiming to provide perspectives for future research.

Author Info: (1) Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China. Department of Research and Development,

Author Info: (1) Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China. Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China. Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China. (2) Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China. Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China. (3) Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China. Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China. (4) Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China. Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China. (5) Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China. Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China. (6) Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China. (7) Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China. Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China.

Improvement in neoantigen prediction via integration of RNA sequencing data for variant calling

INTRODUCTION: Neoantigen-based immunotherapy has emerged as a promising strategy for improving the life expectancy of cancer patients. This therapeutic approach heavily relies on accurate identification of cancer mutations using DNA sequencing (DNAseq) data. However, current workflows tend to provide a large number of neoantigen candidates, of which only a limited number elicit efficient and immunogenic T-cell responses suitable for downstream clinical evaluation. To overcome this limitation and increase the number of high-quality immunogenic neoantigens, we propose integrating RNA sequencing (RNAseq) data into the mutation identification step in the neoantigen prediction workflow. METHODS: In this study, we characterize the mutation profiles identified from DNAseq and/or RNAseq data in tumor tissues of 25 patients with colorectal cancer (CRC). Immunogenicity was then validated by ELISpot assay using long synthesis peptides (sLP). RESULTS: We detected only 22.4% of variants shared between the two methods. In contrast, RNAseq-derived variants displayed unique features of affinity and immunogenicity. We further established that neoantigen candidates identified by RNAseq data significantly increased the number of highly immunogenic neoantigens (confirmed by ELISpot) that would otherwise be overlooked if relying solely on DNAseq data. DISCUSSION: This integrative approach holds great potential for improving the selection of neoantigens for personalized cancer immunotherapy, ultimately leading to enhanced treatment outcomes and improved survival rates for cancer patients.

Author Info: (1) Medical Genetics Institute, Ho Chi Minh, Vietnam. (2) Medical Genetics Institute, Ho Chi Minh, Vietnam. (3) University Medical Center Ho Chi Minh City, Ho Chi Minh, Vietnam. (4

Author Info: (1) Medical Genetics Institute, Ho Chi Minh, Vietnam. (2) Medical Genetics Institute, Ho Chi Minh, Vietnam. (3) University Medical Center Ho Chi Minh City, Ho Chi Minh, Vietnam. (4) Medical Genetics Institute, Ho Chi Minh, Vietnam. (5) Medical Genetics Institute, Ho Chi Minh, Vietnam. (6) Medical Genetics Institute, Ho Chi Minh, Vietnam. (7) University Medical Center Ho Chi Minh City, Ho Chi Minh, Vietnam. (8) Medical Genetics Institute, Ho Chi Minh, Vietnam. (9) University Medical Center Ho Chi Minh City, Ho Chi Minh, Vietnam. (10) University Medical Center Ho Chi Minh City, Ho Chi Minh, Vietnam. (11) University Medical Center Ho Chi Minh City, Ho Chi Minh, Vietnam. (12) Medical Genetics Institute, Ho Chi Minh, Vietnam. (13) Medical Genetics Institute, Ho Chi Minh, Vietnam. (14) Medical Genetics Institute, Ho Chi Minh, Vietnam. (15) Medical Genetics Institute, Ho Chi Minh, Vietnam.

A phase 1 trial of adoptive transfer of vaccine-primed autologous circulating T cells in ovarian cancer

We have previously shown that vaccination with tumor-pulsed dendritic cells amplifies neoantigen recognition in ovarian cancer. Here, in a phase 1 clinical study ( NCT01312376 /UPCC26810) including 19 patients, we show that such responses are further reinvigorated by subsequent adoptive transfer of vaccine-primed, ex vivo-expanded autologous peripheral blood T cells. The treatment is safe, and epitope spreading with novel neopeptide reactivities was observed after cell infusion in patients who experienced clinical benefit, suggesting reinvigoration of tumor-sculpting immunity.

Author Info: (1) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital

Author Info: (1) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (2) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. Center for Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (3) Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA. (4) Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (5) Institute of Pathology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland. (6) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (7) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (8) Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA. (9) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (10) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (11) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (12) Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA. (13) Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA. Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA. (14) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (15) Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece. (16) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland. (17) SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland. Bioinformatics Competence Center, ƒcole Polytechnique FŽdŽrale de Lausanne, Lausanne, Switzerland. (18) Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA. (19) Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (20) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland. (21) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (22) Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA. (23) Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA. (24) Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece. (25) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. Center for Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (26) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. (27) Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Lausanne, Switzerland. george.coukos@chuv.ch. Center for Cell Immunotherapy, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland. george.coukos@chuv.ch.

A H3K27M-targeted vaccine in adults with diffuse midline glioma

Substitution of lysine 27 to methionine in histone H3 (H3K27M) defines an aggressive subtype of diffuse glioma. Previous studies have shown that a H3K27M-specific long peptide vaccine (H3K27M-vac) induces mutation-specific immune responses that control H3K27M(+) tumors in major histocompatibility complex-humanized mice. Here we describe a first-in-human treatment with H3K27M-vac of eight adult patients with progressive H3K27M(+) diffuse midline glioma on a compassionate use basis. Five patients received H3K27M-vac combined with anti-PD-1 treatment based on physician's discretion. Repeat vaccinations with H3K27M-vac were safe and induced CD4(+) T cell-dominated, mutation-specific immune responses in five of eight patients across multiple human leukocyte antigen types. Median progression-free survival after vaccination was 6.2 months and median overall survival was 12.8 months. One patient with a strong mutation-specific T cell response after H3K27M-vac showed pseudoprogression followed by sustained complete remission for >31 months. Our data demonstrate safety and immunogenicity of H3K27M-vac in patients with progressive H3K27M(+) diffuse midline glioma.

Author Info: (1) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelb

Author Info: (1) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany. DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany. (2) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Immune Monitoring Unit, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany. (3) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Immune Monitoring Unit, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany. (4) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany. DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany. (5) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany. DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany. (6) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Immune Monitoring Unit, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany. Helmholtz Institute for Translational Oncology (HI-TRON) Mainz, German Cancer Research Center, Mainz, Germany. (7) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany. DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany. (8) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany. DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany. (9) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Immune Monitoring Unit, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany. (10) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. Immune Monitoring Unit, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany. (11) Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany. National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany. (12) Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany. DKTK Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany. (13) Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany. (14) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany. Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany. (15) Department of Neurosurgery, CharitŽ-UniversitŠtsmedizin Berlin, Berlin, Germany. (16) Department of Neurology with Institute of Translational Neurology, University of MŸnster, MŸnster, Germany. (17) Division of Clinical Neurooncology, Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany. (18) Department of Neurosurgery, University of Munich LMU, Munich, Germany. (19) Institute of Cell Biology, Department of Immunology, University of TŸbingen, TŸbingen, Germany. (20) Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany. DKTK Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany. (21) Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany. (22) Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany. DKTK Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany. (23) Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany. National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany. (24) Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany. National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany. (25) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. m.platten@dkfz-heidelberg.de. Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany. m.platten@dkfz-heidelberg.de. DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany. m.platten@dkfz-heidelberg.de. Immune Monitoring Unit, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany. m.platten@dkfz-heidelberg.de. Helmholtz Institute for Translational Oncology (HI-TRON) Mainz, German Cancer Research Center, Mainz, Germany. m.platten@dkfz-heidelberg.de. (26) DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany. k.sahm@dkfz-heidelberg.de. Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany. k.sahm@dkfz-heidelberg.de. DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany. k.sahm@dkfz-heidelberg.de.

Manipulating mitochondrial electron flow enhances tumor immunogenicity

Although tumor growth requires the mitochondrial electron transport chain (ETC), the relative contribution of complex I (CI) and complex II (CII), the gatekeepers for initiating electron flow, remains unclear. In this work, we report that the loss of CII, but not that of CI, reduces melanoma tumor growth by increasing antigen presentation and T cell-mediated killing. This is driven by succinate-mediated transcriptional and epigenetic activation of major histocompatibility complex-antigen processing and presentation (MHC-APP) genes independent of interferon signaling. Furthermore, knockout of methylation-controlled J protein (MCJ), to promote electron entry preferentially through CI, provides proof of concept of ETC rewiring to achieve antitumor responses without side effects associated with an overall reduction in mitochondrial respiration in noncancer cells. Our results may hold therapeutic potential for tumors that have reduced MHC-APP expression, a common mechanism of cancer immunoevasion.

Author Info: (1) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (2) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (3) Salk Institute for Biological Studies, L

Author Info: (1) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (2) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (3) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (4) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (5) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (6) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (7) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA. (8) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (9) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA. (10) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (11) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (12) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (13) Whitehead Institute Metabolomics Core Facility, Cambridge, MA 02139, USA. (14) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (15) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (16) Department of Immunology and Microbiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA. (17) Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA. (18) Departments of Pathology, Dermatology, and Immunology, Yale University School of Medicine, New Haven, CT 06520, USA. (19) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (20) Salk Institute for Biological Studies, La Jolla, CA 92037, USA. (21) Salk Institute for Biological Studies, La Jolla, CA 92037, USA.

Responses of patients with cancer to mRNA vaccines depend on the time interval between vaccination and last treatment

BACKGROUND: Personalized mRNA vaccines are promising new therapeutic options for patients with cancer. Because mRNA vaccines are not yet approved for first-line therapy, the vaccines are presently applied to individuals that received prior therapies that can have immunocompromising effects. There is a need to address how prior treatments impact mRNA vaccine outcomes. METHOD: Therefore, we analyzed the response to BioNTech/Pfizer's anti-SARS-CoV-2 mRNA vaccine in 237 oncology outpatients, which cover a broad spectrum of hematologic malignancies and solid tumors and a variety of treatments. Patients were stratified by the time interval between the last treatment and first vaccination and by the presence or absence of florid tumors and IgG titers and T cell responses were analyzed 14 days after the second vaccination. RESULTS: Regardless of the last treatment time point, our data indicate that vaccination responses in patients with checkpoint inhibition were comparable to healthy controls. In contrast, patients after chemotherapy or cortisone therapy did not develop an immune response until 6 months after the last systemic therapy and patients after Cht-immune checkpoint inhibitor and tyrosine kinase inhibitor therapy only after 12 months. CONCLUSION: Accordingly, our data support that timing of mRNA-based therapy is critical and we suggest that at least a 6-months or 12-months waiting interval should be observed before mRNA vaccination in systemically treated patients.

Author Info: (1) Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany. (2) Department of Otorhinolaryngology, Technical University of Munich, Munich,

Author Info: (1) Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany. (2) Department of Otorhinolaryngology, Technical University of Munich, Munich, Germany. (3) MVZ Laboratory, Freising, Germany. (4) Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany. (5) Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany. (6) Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany. (7) Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany. (8) System Technologies and Image Exploitation IOSB, Fraunhofer Institute of Optronics, Karlsruhe, Germany. (9) Division of Applied Health and Social Sciences, Technical University of Applied Sciences, Rosenheim, Germany. (10) MVZ Laboratory, Freising, Germany. (11) Oncological Center Donauwšrth, Donauwšrth, Germany dietmar.zehn@tum.de Louisa.hempel@sfu.ac.at dirk.hempel@gmail.com. (12) Sigmund Freud Medical University, Vienna, Austria dietmar.zehn@tum.de Louisa.hempel@sfu.ac.at dirk.hempel@gmail.com. (13) Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany dietmar.zehn@tum.de Louisa.hempel@sfu.ac.at dirk.hempel@gmail.com.

Personalized neoantigen cancer vaccines: An analysis of the clinical and commercial potential of ongoing development programs

Neoantigen cancer vaccines harbor promise as next-generation immuno-oncology therapies, whereby cancer vaccines are tailored to the patient's tumor antigen and represent the future of personalized cancer therapy. While several biotech companies have ongoing development programs, little has been published about the true commercial potential of these innovative therapies and the challenges these products will face upon regulatory approval. In this paper, we provide an overview of neoantigen cancer vaccine development programs and discuss the commercial environment these therapies will face upon launch.

Author Info: (1) EY-Parthenon, Basel, Switzerland. Electronic address: Maurice.perrinjaquet@parthenon.ey.com. (2) EY-Parthenon, Berlin, Germany.

Author Info: (1) EY-Parthenon, Basel, Switzerland. Electronic address: Maurice.perrinjaquet@parthenon.ey.com. (2) EY-Parthenon, Berlin, Germany.

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.