Antigens in Cancer

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Immunology and Immunotherapy".

Deadline for manuscript submissions: closed (20 August 2023) | Viewed by 10928

Special Issue Editor


E-Mail Website
Guest Editor
Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
Interests: CAR T cell therapy; thyroid cancer

Special Issue Information

Dear Colleagues,

The surface antigens which are restricted or more abundant in cancer cells can be exploited to deliver cytotoxic drugs, biologics, and immune cells to tumor cells. Tumor antigens can be either unique to tumor cells (tumor-specific antigens, TSAs) or relatively more abundant (tumor associated antigens, TAAs). TSAs can be created by the presence of new genetic material of viral origin (e.g., human papillomavirus) or by neoantigens generated by the inherent genetic instability of cancer cells. TAAs are present in normal cells but become abnormally overexpressed in tumor cells (e.g., PSMA, mesothelin, ICAM-1 or Her2) or those that are expressed primarily during embryonic development or are otherwise tissue-restricted (such as cancer testis antigens). TSAs have been more commonly identified as peptide epitopes in complex with MHC, although some neoantigens in common surface molecules have also been identified (e.g., EGFRvIII). All of these can be exploited for targeted therapies.

The types of drugs that are designed to be specific to tumor antigens are quite diverse. For example, drugs based on the radioisotope 177Lutetium chelated to peptides specific to SSTR2 and PSMA have been approved as cancer therapy against SSTR2-positive gastroenteropancreatic/neuroendocrine cancers and metastatic castration-resistant prostate cancer, respectively. Antibodies are a natural choice to target tumor surface antigens, as these can kill tumor cells via various mechanisms involving natural killer cells, macrophages, complement activation or by directly blocking cell signaling. To further enhance antibody mediated killing, cytotoxic drugs conjugated to antibodies have been developed and proven efficacious, (e.g., T-DM1 (trastuzumab emtansine)).

Recently, T-cell immunotherapy has emerged as a powerful tool for personalized medicine, where autologous or allogeneic T cells are genetically modified to target tumor antigens by the expression of TCRs specific to tumor peptide/MHC complexes or by chimeric antigen receptors (CARs). However, the success of targeting a TAA for one drug type does not guarantee that it can be used for other types of drugs. For example, PSMA-specific radiotherapy (177Lu-PSMA-617) has proven to be a highly effective drug with limited side effects. In contrast, recent trials with CAR T cells targeting PSMA reported multiple patient deaths, implying that the threshold of antigen density eliciting toxicity can be quite different depending on the type of treatment.

In this special issue, we are particularly interested in papers that report on the identification and/or characterization of novel antigens in cancer and different agents that target these novel antigens. Moreover, papers leading to the development of specific methodologies aiming at the better screening/identification/quantification of cancer antigens would be of great interest. Reviews that summarize current knowledge gained from the preclinical and clinical studies of TCR- and CAR-T cells against cancer would be particularly useful. Potential topics include, but are not limited to:

  • Identification or prioritization of novel cancer antigens
  • Methodologies for the detection of cancer antigens
  • Next-generation or fine-tuning of TCR or CAR constructs to overcome immune suppression or avoid toxicity
  • Novel cancer-antigen-specific small molecules or peptides as carriers of cytotoxic drugs or radioisotopes
  • Novel antibody designs and formats for improved pharmacokinetics or conjugation with cytotoxic drugs or radioisotopes.

Prof. Dr. Moonsoo Jin
Guest Editor

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. Cancers is an international peer-reviewed open access semimonthly 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 2900 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

  • tumor antigens
  • immunotherapy
  • chimeric antigen receptor (CAR)
  • antibody conjugates
  • drug carriers

Published Papers (4 papers)

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

Research

Jump to: Review

20 pages, 7360 KiB  
Article
Chimeric Antigen Receptor T Cell Therapy Targeting Epithelial Cell Adhesion Molecule in Gastric Cancer: Mechanisms of Tumor Resistance
by Yanping Yang, Raymond Louie, Janusz Puc, Yogindra Vedvyas, Yago Alcaina, Irene M. Min, Matt Britz, Fabio Luciani and Moonsoo M. Jin
Cancers 2023, 15(23), 5552; https://doi.org/10.3390/cancers15235552 - 23 Nov 2023
Cited by 1 | Viewed by 1208
Abstract
Epithelial cell adhesion molecule (EpCAM) is a tumor-associated antigen that is frequently overexpressed in various carcinomas. We have developed chimeric antigen receptor (CAR) T cells specifically targeting EpCAM for the treatment of gastric cancer. This study sought to unravel the precise mechanisms by [...] Read more.
Epithelial cell adhesion molecule (EpCAM) is a tumor-associated antigen that is frequently overexpressed in various carcinomas. We have developed chimeric antigen receptor (CAR) T cells specifically targeting EpCAM for the treatment of gastric cancer. This study sought to unravel the precise mechanisms by which tumors evade immune surveillance and develop resistance to CAR T cell therapy. Through a combination of whole-body CAR T cell imaging and single-cell multiomic analyses, we uncovered intricate interactions between tumors and tumor-infiltrating lymphocytes (TILs). In a gastric cancer model, tumor-infiltrating CD8 T cells exhibited both cytotoxic and exhausted phenotypes, while CD4 T cells were mainly regulatory T cells. A T cell receptor (TCR) clonal analysis provided evidence of CAR T cell proliferation and clonal expansion within resistant tumors, which was substantiated by whole-body CAR T cell imaging. Furthermore, single-cell transcriptomics showed that tumor cells in mice with refractory or relapsing outcomes were enriched for genes involved in major histocompatibility complex (MHC) and antigen presentation pathways, interferon-γ and interferon-α responses, mitochondrial activities, and a set of genes (e.g., CD74, IDO1, IFI27) linked to tumor progression and unfavorable disease prognoses. This research highlights an approach that combines imaging and multiomic methodologies to concurrently characterize the evolution of tumors and the differentiation of CAR T cells. Full article
(This article belongs to the Special Issue Antigens in Cancer)
Show Figures

Figure 1

14 pages, 3672 KiB  
Article
Preclinical Assessment of Immunogenicity and Protectivity of Novel ROR1 Fusion Proteins in a Mouse Tumor Model
by Hadi Hassannia, Mohammad Mehdi Amiri, Mojgan Ghaedi, Ramezan-Ali Sharifian, Forough Golsaz-Shirazi, Mahmood Jeddi-Tehrani and Fazel Shokri
Cancers 2022, 14(23), 5827; https://doi.org/10.3390/cancers14235827 - 26 Nov 2022
Cited by 2 | Viewed by 1350
Abstract
The receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a new tumor associated antigen (TAA) which is overexpressed in several hematopoietic and solid malignancies. The present study aimed to produce and evaluate different fusion proteins of mouse ROR1 (mROR1) to enhance immunogenicity and [...] Read more.
The receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a new tumor associated antigen (TAA) which is overexpressed in several hematopoietic and solid malignancies. The present study aimed to produce and evaluate different fusion proteins of mouse ROR1 (mROR1) to enhance immunogenicity and protective efficacy of ROR1. Four ROR1 fusion proteins composed of extracellular region of mROR1, immunogenic fragments of TT as well as Fc region of mouse IgG2a were produced and employed to immunize Balb/C mice. Humoral and cellular immune responses and anti-tumor effects of these fusion proteins were evaluated using two different syngeneic murine ROR1+ tumor models. ROR1-specific antibodies were induced in all groups of mice. The levels of IFN-γ, IL-17 and IL-22 cytokines in culture supernatants of stimulated splenocytes were increased in all groups of immunized mice, particularly mice immunized with TT-mROR1-Fc fusion proteins. The frequency of ROR1-specific CTLs was higher in mice immunized with TT-mROR1-Fc fusion proteins. Finally, results of tumor challenge in immunized mice showed that immunization with TT-mROR1-Fc fusion proteins completely inhibited ROR1+ tumor cells growth in two different syngeneic tumor models until day 120 post tumor challenge. Our preclinical findings, for the first time, showed that our fusion proteins could be considered as a potential candidate vaccine for active immunotherapy of ROR1-expressing malignancies. Full article
(This article belongs to the Special Issue Antigens in Cancer)
Show Figures

Figure 1

18 pages, 2217 KiB  
Article
A Novel Proteogenomic Integration Strategy Expands the Breadth of Neo-Epitope Sources
by Haitao Xiang, Le Zhang, Fanyu Bu, Xiangyu Guan, Lei Chen, Haibo Zhang, Yuntong Zhao, Huanyi Chen, Weicong Zhang, Yijian Li, Leo Jingyu Lee, Zhanlong Mei, Yuan Rao, Ying Gu, Yong Hou, Feng Mu and Xuan Dong
Cancers 2022, 14(12), 3016; https://doi.org/10.3390/cancers14123016 - 19 Jun 2022
Cited by 2 | Viewed by 2339
Abstract
Tumor-specific antigens can activate T cell-based antitumor immune responses and are ideal targets for cancer immunotherapy. However, their identification is still challenging. Although mass spectrometry can directly identify human leukocyte antigen (HLA) binding peptides in tumor cells, it focuses on tumor-specific antigens derived [...] Read more.
Tumor-specific antigens can activate T cell-based antitumor immune responses and are ideal targets for cancer immunotherapy. However, their identification is still challenging. Although mass spectrometry can directly identify human leukocyte antigen (HLA) binding peptides in tumor cells, it focuses on tumor-specific antigens derived from annotated protein-coding regions constituting only 1.5% of the genome. We developed a novel proteogenomic integration strategy to expand the breadth of tumor-specific epitopes derived from all genomic regions. Using the colorectal cancer cell line HCT116 as a model, we accurately identified 10,737 HLA-presented peptides, 1293 of which were non-canonical peptides that traditional database searches could not identify. Moreover, we found eight tumor neo-epitopes derived from somatic mutations, four of which were not previously reported. Our findings suggest that this new proteogenomic approach holds great promise for increasing the number of tumor-specific antigen candidates, potentially enlarging the tumor target pool and improving cancer immunotherapy. Full article
(This article belongs to the Special Issue Antigens in Cancer)
Show Figures

Figure 1

Review

Jump to: Research

28 pages, 1893 KiB  
Review
Targeting CD38 in Neoplasms and Non-Cancer Diseases
by Wojciech Szlasa, Jakub Czarny, Natalia Sauer, Katarzyna Rakoczy, Natalia Szymańska, Jakub Stecko, Maksymilian Kołodziej, Maciej Kaźmierczak and Ewa Barg
Cancers 2022, 14(17), 4169; https://doi.org/10.3390/cancers14174169 - 28 Aug 2022
Cited by 11 | Viewed by 5343
Abstract
CD38 is a myeloid antigen present both on the cell membrane and in the intracellular compartment of the cell. Its occurrence is often enhanced in cancer cells, thus making it a potential target in anticancer therapy. Daratumumab and isatuximab already received FDA approval, [...] Read more.
CD38 is a myeloid antigen present both on the cell membrane and in the intracellular compartment of the cell. Its occurrence is often enhanced in cancer cells, thus making it a potential target in anticancer therapy. Daratumumab and isatuximab already received FDA approval, and novel agents such as MOR202, TAK079 and TNB-738 undergo clinical trials. Also, novel therapeutics such as SAR442085 aim to outrank the older antibodies against CD38. Multiple myeloma and immunoglobulin light-chain amyloidosis may be effectively treated with anti-CD38 immunotherapy. Its role in other hematological malignancies is also important concerning both diagnostic process and potential treatment in the future. Aside from the hematological malignancies, CD38 remains a potential target in gastrointestinal, neurological and pulmonary system disorders. Due to the strong interaction of CD38 with TCR and CD16 on T cells, it may also serve as the biomarker in transplant rejection in renal transplant patients. Besides, CD38 finds its role outside oncology in systemic lupus erythematosus and collagen-induced arthritis. CD38 plays an important role in viral infections, including AIDS and COVID-19. Most of the undergoing clinical trials focus on the use of anti-CD38 antibodies in the therapy of multiple myeloma, CD19- B-cell malignancies, and NK cell lymphomas. This review focuses on targeting CD38 in cancer and non-cancerous diseases using antibodies, cell-based therapies and CD38 inhibitors. We also provide a summary of current clinical trials targeting CD38. Full article
(This article belongs to the Special Issue Antigens in Cancer)
Show Figures

Figure 1

Back to TopTop