Vaccines Targeting the Tumor Microenvironment: Challenges and Future Prospects

A special issue of Vaccines (ISSN 2076-393X). This special issue belongs to the section "Cancer Vaccines and Immunotherapy".

Deadline for manuscript submissions: 30 November 2024 | Viewed by 696

Special Issue Editors


E-Mail Website
Guest Editor
1. Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
2. UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
Interests: angiogenesis; cancer immunology; cancer immunotherapy; cancer vaccines; CD8+ T cells; chemokines; dendritic cells; pericytes; tertiary lymphoid structures; tumor microenvironment; vascular endothelial cells

E-Mail Website
Guest Editor
Department Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
Interests: tumor angiogenesis; cancer immunology; cancer immunotherapy; cancer vaccines; antibodies

Special Issue Information

Dear Colleagues,

Immunotherapies integrating immune checkpoint inhibitors (ICI) represent the current standard of care in the setting of many forms of solid cancer, yet they effectively treat only a minority of patients and are leveraged by the prerequisite for the presence of tumor-reactive T (or B) cells in the patient at baseline. To augment levels of such therapeutic immune effector cells, active vaccination approaches are warranted as priming regimens prior to the administration of ICI or for inclusion in combination intervention protocols to improved patient outcomes. Although cancer vaccines have traditionally targeted tumor cell-associated antigens (including mutated neoantigens), the tumor microenvironment can enforce altered epigenetic programming in its component (non-tumor) stromal cell populations including cancer-associated fibroblasts (CAFs), vascular pericytes/mural cells, and vascular endothelial cells, among others, providing a diverse array of TME-associated antigens capable of being differentially recognized vs. normal tissues by the adaptive immune system. In this Special Issue, we welcome the submission of articles focused on cutting-edge vaccine-based approaches to improve the host response to cancer when applied as monotherapies or in combination with alternate immunotherapies such as ICI, TME conditioning agents, adoptive (T/CAR-T) cell therapies, or others.

Prof. Dr. Walter J. Storkus
Dr. Devin Lowe
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Vaccines is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • vaccine
  • adoptive cell therapy
  • CAR-T cells
  • immune checkpoint inhibitors
  • immunotherapy
  • stroma
  • tumor microenvironment

Published Papers (1 paper)

Order results
Result details
Select all
Export citation of selected articles as:

Research

15 pages, 3867 KiB  
Article
STAT1-Deficient HPV E6/E7-Associated Cancers Maintain Host Immunocompetency against Therapeutic Intervention
by Ling Lim, Ming-Hung Hu, Darrell Fan, Hsin-Fang Tu, Ya-Chea Tsai, Michelle Cheng, Suyang Wang, Chih-Long Chang, Tzyy-Choou Wu and Chien-Fu Hung
Vaccines 2024, 12(4), 430; https://doi.org/10.3390/vaccines12040430 - 17 Apr 2024
Viewed by 400
Abstract
Human papillomavirus (HPV) remains a global health concern because it contributes to the initiation of various HPV-associated cancers such as anal, cervical, oropharyngeal, penile, vaginal, and vulvar cancer. In HPV-associated cancers, oncogenesis begins with an HPV infection, which is linked to the activation [...] Read more.
Human papillomavirus (HPV) remains a global health concern because it contributes to the initiation of various HPV-associated cancers such as anal, cervical, oropharyngeal, penile, vaginal, and vulvar cancer. In HPV-associated cancers, oncogenesis begins with an HPV infection, which is linked to the activation of the Janus protein tyrosine kinase (JAK)/STAT signaling pathway. Various STAT signaling pathways, such as STAT3 activation, have been well documented for their tumorigenic role, yet the role of STAT1 in tumor formation remains unclear. In the current study, STAT1−/− mice were used to investigate the role of STAT1 in the tumorigenesis of a spontaneous HPV E6/E7-expressing oral tumor model. Subsequently, our candidate HPV DNA vaccine CRT/E7 was administered to determine whether the STAT1−/− host preserves a therapeutic-responsive tumor microenvironment. The results indicated that STAT1−/− induces robust tumorigenesis, yet a controlled tumor response was attained upon CRT/E7 vaccination. Characterizing this treatment effect, immunological analysis found a higher percentage of circulating CD4+ and CD8+ T cells and tumor-specific cytotoxic T cells. In addition, a reduction in exhaustive lymphocyte activity was observed. Further analysis of a whole-cell tumor challenge affirmed these findings, as spontaneous tumor growth was more rapid in STAT1−/− mice. In conclusion, STAT1 deletion accelerates tumorigenesis, but STAT1−/− mice maintains immunocompetency in CRT/E7 treatments. Full article
Show Figures

Figure 1

Back to TopTop