4.8 Article

Expanding cross-presenting dendritic cells enhances oncolytic virotherapy and is critical for long-term anti-tumor immunity

期刊

NATURE COMMUNICATIONS
卷 13, 期 1, 页码 -

出版社

NATURE PORTFOLIO
DOI: 10.1038/s41467-022-34791-8

关键词

-

资金

  1. Swedish Research Council [2017-00565]
  2. Swedish Society for Medical Research, SSMF [P16-0026]
  3. National Institutes of Health (NIH) [R01CA229818]
  4. NIH [R01CA229818, R01CA257195, R37 CA246239]
  5. Alliance for Cancer Gene Therapy
  6. Applebaum Foundation
  7. Cancer Research Institute (CRI) Lloyd Old STAR Award
  8. Damon Runyon Cancer Research Foundation Clinical Investigator Award
  9. Swedish Research Council [2017-00565] Funding Source: Swedish Research Council

向作者/读者索取更多资源

Expanding dendritic cells to increase tumor antigen cross-presentation improves oncolytic virotherapy and induces neoepitope-specific T cell response.
Immunotherapies directly enhancing anti-tumor CD8(+) T cell responses have yielded measurable but limited success, highlighting the need for alternatives. Anti-tumor T cell responses critically depend on antigen presenting dendritic cells (DC), and enhancing mobilization, antigen loading and activation of these cells represent an attractive possibility to potentiate T cell based therapies. Here we show that expansion of DCs by Flt3L administration impacts in situ vaccination with oncolytic Newcastle Disease Virus (NDV). Mechanistically, NDV activates DCs and sensitizes them to dying tumor cells through upregulation of dead-cell receptors and synergizes with Flt3L to promote anti-tumor CD8(+) T cell cross-priming. In vivo, Flt3L-NDV in situ vaccination induces parallel amplification of virus- and tumor-specific T cells, including CD8(+) T cells reactive to newly-described neoepitopes, promoting long-term tumor control. Cross-presenting conventional Type 1 DCs are indispensable for the anti-tumor, but not anti-viral, T cell response, and type I IFN-dependent CD4(+) Th1 effector cells contribute to optimal anti-tumor immunity. These data demonstrate that mobilizing DCs to increase tumor antigen cross-presentation improves oncolytic virotherapy and that neoepitope-specific T cells can be induced without individualized, ex vivo manufactured vaccines. Strategies to advance T cell based immune therapies are mostly focusing on the improvement of CD8 T cell effector functions, such as cytotoxicity or recruitment to the tumor. Here authors show that by combining in situ vaccination with oncolytic Newcastle Disease Virus and Flt3L-driven dendritic cell expansion, the anti-tumor T cell response is amplified via increased antigen cross-presentation.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.8
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

Review Oncology

Pathogen Molecular Pattern Receptor Agonists: Treating Cancer by Mimicking Infection

Mark Aleynick, Judit Svensson-Arvelund, Christopher R. Flowers, Aurelien Marabelle, Joshua D. Brody

CLINICAL CANCER RESEARCH (2019)

Article Biochemistry & Molecular Biology

Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination

Linda Hammerich, Thomas U. Marron, Ranjan Upadhyay, Judit Svensson-Arvelund, Maxime Dhainaut, Shafinaz Hussein, Yougen Zhan, Dana Ostrowski, Michael Yellin, Henry Marsh, Andres M. Salazar, Adeeb H. Rahman, Brian D. Brown, Miriam Merad, Joshua D. Brody

NATURE MEDICINE (2019)

Article Immunology

A HER2-Displaying Virus-Like Particle Vaccine Protects from Challenge with Mammary Carcinoma Cells in a Mouse Model

Lisa Nika, Sara Cuadrado-Castano, Guha Asthagiri Arunkumar, Clemens Gruenwald-Gruber, Meagan McMahon, Krisztina Koczka, Adolfo Garcia-Sastre, Florian Krammer, Reingard Grabherr

VACCINES (2019)

Article Oncology

Antitumor T-cell Homeostatic Activation Is Uncoupled from Homeostatic Inhibition by Checkpoint Blockade

Netonia Marshall, Keino Hutchinson, Thomas U. Marron, Mark Aleynick, Linda Hammerich, Ranjan Upadhyay, Judit Svensson-Arvelund, Brian D. Brown, Miriam Merad, Joshua D. Brody

CANCER DISCOVERY (2019)

Article Cell Biology

Newcastle Disease Virus (NDV) Oncolytic Activity in Human Glioma Tumors Is Dependent on CDKN2A-Type I IFN Gene Cluster Codeletion

Noemi Garcia-Romero, Irina Palacin-Aliana, Susana Esteban-Rubio, Rodrigo Madurga, Sergio Rius-Rocabert, Josefa Carrion-Navarro, Jesus Presa, Sara Cuadrado-Castano, Pilar Sanchez-Gomez, Adolfo Garcia-Sastre, Estanislao Nistal-Villan, Angel Ayuso-Sacido

Article Oncology

A Critical Role for Fas-Mediated Off-Target Tumor Killing in T-cell Immunotherapy

Ranjan Upadhyay, Jonathan A. Boiarsky, Gvantsa Pantsulaia, Judit Svensson-Arvelund, Matthew J. Lin, Aleksandra Wroblewska, Sherry Bhalla, Nathalie Scholler, Adrian Bot, John M. Rossi, Norah Sadek, Samir Parekh, Alessandro Lagana, Alessia Baccarini, Miriam Merad, Brian D. Brown, Joshua D. Brody

Summary: The study revealed the crucial role of Fas-FasL in T cell killing of tumors and the bystander killing of antigen-negative tumor cells. This new finding may enhance the efficacy of immunotherapies and prevent cancer relapse by targeting both antigen expression and antigen-loss variants.

CANCER DISCOVERY (2021)

Review Immunology

Engineering Non-Human RNA Viruses for Cancer Therapy

Vicent Tur-Planells, Adolfo Garcia-Sastre, Sara Cuadrado-Castano, Estanislao Nistal-Villan

Summary: This review provides an overview of non-human-adapted RNA viruses used in cancer therapy and the diverse strategies employed to optimize their therapeutic capabilities. Through this article, we gain insights into the promising advancements in virotherapy and its potential to revolutionize cancer treatment.

VACCINES (2023)

Article Oncology

Avian Paramyxovirus 4 Antitumor Activity Leads to Complete Remissions and Long-term Protective Memory in Preclinical Melanoma and Colon Carcinoma Models

Aryana Javaheri, Yonina Bykov, Ignacio Mena, Adolfo Garcia-Sastre, Sara Cuadrado-Castano

Summary: The study highlights the promising oncolytic properties of APMV-4 Duck/Hong Kong/D3/1975 virus in murine colon carcinoma and melanoma tumor models, surpassing the therapeutic potential of NDV. Additionally, recombinant APMV-4 viruses have demonstrated significant antitumor capabilities, paving the way for the development of next-generation viral therapeutics.

CANCER RESEARCH COMMUNICATIONS (2022)

暂无数据