Cancers Precision Immunotherapy

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

Deadline for manuscript submissions: 31 July 2024 | Viewed by 12961

Special Issue Editor


grade E-Mail Website
Guest Editor
Froedtert & The Medical College of Wisconsin Clinical Cancer Center, Milwaukee, WI 53226, USA
Interests: rare diseases; immunotherapy; precision oncology
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Precision immunotherapy refers to the field of matching immune therapies to a tumor's immunomic landscape in order for that tumor to be treated with immunotherapy in a "precise" manner. Because the immune environment of each tumor may be distinct, treating precisely appears to require a personalized/individualized immunotherapy approach. There are many innovative ways to address the immune deficits that allow tumors to grow. Some of the new and powerful tools in the therapeutic armamentarium include CAR T-cells, bispecific antibodies, personalized vaccines, immune checkpoint inhibitors and immune stimulatory molecules. Interrogating the tumor immunogram and matching the patient to the compounds that are most likely to reactivate the immune system and permit it to eliminate cancer cells requires multiple advanced molecular technologies. Even so, precision immunotherapy has shown a remarkable ability to eradicate even advanced malignancies.

Prof. Dr. Razelle Kurzrock
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

  • immunotherapy
  • checkpoint blockade
  • CAR T cells
  • bispecific antibodies
  • personalized vaccines

Published Papers (5 papers)

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

Research

Jump to: Review

21 pages, 2296 KiB  
Article
Inflammation and Immunity Gene Expression Patterns and Machine Learning Approaches in Association with Response to Immune-Checkpoint Inhibitors-Based Treatments in Clear-Cell Renal Carcinoma
by Nikolas Dovrolis, Hector Katifelis, Stamatiki Grammatikaki, Roubini Zakopoulou, Aristotelis Bamias, Michalis V. Karamouzis, Kyriakos Souliotis and Maria Gazouli
Cancers 2023, 15(23), 5637; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15235637 - 29 Nov 2023
Viewed by 1260
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common renal cancer. Despite the rapid evolution of targeted therapies, immunotherapy with checkpoint inhibition (ICI) as well as combination therapies, the cure of metastatic ccRCC (mccRCC) is infrequent, while the optimal use of the [...] Read more.
Clear cell renal cell carcinoma (ccRCC) is the most common renal cancer. Despite the rapid evolution of targeted therapies, immunotherapy with checkpoint inhibition (ICI) as well as combination therapies, the cure of metastatic ccRCC (mccRCC) is infrequent, while the optimal use of the various novel agents has not been fully clarified. With the different treatment options, there is an essential need to identify biomarkers to predict therapeutic efficacy and thus optimize therapeutic approaches. This study seeks to explore the diversity in mRNA expression profiles of inflammation and immunity-related circulating genes for the development of biomarkers that could predict the effectiveness of immunotherapy-based treatments using ICIs for individuals with mccRCC. Gene mRNA expression was tested by the RT2 profiler PCR Array on a human cancer inflammation and immunity crosstalk kit and analyzed for differential gene expression along with a machine learning approach for sample classification. A number of mRNAs were found to be differentially expressed in mccRCC with a clinical benefit from treatment compared to those who progressed. Our results indicate that gene expression can classify these samples with high accuracy and specificity. Full article
(This article belongs to the Special Issue Cancers Precision Immunotherapy)
Show Figures

Figure 1

21 pages, 3896 KiB  
Article
Defining the Immune Checkpoint Landscape in Human Colorectal Cancer Highlights the Relevance of the TIGIT/CD155 Axis for Optimizing Immunotherapy
by Kathleen Ducoin, Linda Bilonda-Mutala, Cécile Deleine, Romain Oger, Emilie Duchalais, Nicolas Jouand, Céline Bossard, Anne Jarry and Nadine Gervois-Segain
Cancers 2022, 14(17), 4261; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14174261 - 31 Aug 2022
Cited by 3 | Viewed by 2085
Abstract
While immune checkpoint (IC) therapies, particularly those targeting the PD-1/PD-L1 axis, have revolutionized the treatment of melanoma and several other cancers, their effect remains very limited in colorectal cancer (CRC). To define a comprehensive landscape of ICs in the human CRC tumor microenvironment [...] Read more.
While immune checkpoint (IC) therapies, particularly those targeting the PD-1/PD-L1 axis, have revolutionized the treatment of melanoma and several other cancers, their effect remains very limited in colorectal cancer (CRC). To define a comprehensive landscape of ICs in the human CRC tumor microenvironment (TME), we evaluated, using multiparametric flow cytometry, their ex vivo expression via tumor-infiltrating lymphocytes (TILs) (n = 40 CRCs) as well as that of their respective ligands on tumor and myeloid cells (n = 29). Supervised flow cytometry analyses showed that (i) most CD3+ TILs expressed PD-1 and TIGIT and, to a lesser extent, Tim-3, Lag3 and NKG2A, and (ii) EpCAM+ tumor cells and CD11b+ myeloid cells differed in their IC ligand expression profile, with a strikingly high expression of CD155 by tumor cells. An in situ analysis of IC and their ligands using immunohistochemistry on paraffin sections of CRC confirmed the overexpression of TIGIT and its ligand, CD155, in the TME. Most interestingly, an unsupervised clustering analysis of IC co-expression on CD4+ and CD8+ TILs identified two tumor subgroups, named IChigh and IClow. Altogether, our findings highlight the TIGIT/CD155 axis as a potential target that could be used in combination IC therapy in CRC. Full article
(This article belongs to the Special Issue Cancers Precision Immunotherapy)
Show Figures

Graphical abstract

Review

Jump to: Research

17 pages, 1185 KiB  
Review
Post-Translational Modifications in Tumor-Associated Antigens as a Platform for Novel Immuno-Oncology Therapies
by Anurag Kumar Srivastava, Giorgia Guadagnin, Paola Cappello and Francesco Novelli
Cancers 2023, 15(1), 138; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15010138 - 26 Dec 2022
Cited by 13 | Viewed by 2821
Abstract
Post-translational modifications (PTMs) are generated by adding small chemical groups to amino acid residues after the translation of proteins. Many PTMs have been reported to correlate with tumor progression, growth, and survival by modifying the normal functions of the protein in tumor cells. [...] Read more.
Post-translational modifications (PTMs) are generated by adding small chemical groups to amino acid residues after the translation of proteins. Many PTMs have been reported to correlate with tumor progression, growth, and survival by modifying the normal functions of the protein in tumor cells. PTMs can also elicit humoral and cellular immune responses, making them attractive targets for cancer immunotherapy. This review will discuss how the acetylation, citrullination, and phosphorylation of proteins expressed by tumor cells render the corresponding tumor-associated antigen more antigenic and affect the immune response in multiple cancers. In addition, the role of glycosylated protein mucins in anti-cancer immunotherapy will be considered. Mucin peptides in combination with stimulating adjuvants have, in fact, been utilized to produce anti-tumor antibodies and vaccines. Finally, we will also outline the results of the clinical trial exploiting glycosylated-MUC1 as a vaccine in different cancers. Overall, PTMs in TAAs could be considered in future therapies to result in lasting anti-tumor responses. Full article
(This article belongs to the Special Issue Cancers Precision Immunotherapy)
Show Figures

Graphical abstract

23 pages, 2801 KiB  
Review
Bone Marrow Immune Microenvironment in Myelodysplastic Syndromes
by Olga Kouroukli, Argiris Symeonidis, Periklis Foukas, Myrto-Kalliopi Maragkou and Eleni P. Kourea
Cancers 2022, 14(22), 5656; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14225656 - 17 Nov 2022
Cited by 9 | Viewed by 2493
Abstract
The BM, the major hematopoietic organ in humans, consists of a pleiomorphic environment of cellular, extracellular, and bioactive compounds with continuous and complex interactions between them, leading to the formation of mature blood cells found in the peripheral circulation. Systemic and local inflammation [...] Read more.
The BM, the major hematopoietic organ in humans, consists of a pleiomorphic environment of cellular, extracellular, and bioactive compounds with continuous and complex interactions between them, leading to the formation of mature blood cells found in the peripheral circulation. Systemic and local inflammation in the BM elicit stress hematopoiesis and drive hematopoietic stem cells (HSCs) out of their quiescent state, as part of a protective pathophysiologic process. However, sustained chronic inflammation impairs HSC function, favors mutagenesis, and predisposes the development of hematologic malignancies, such as myelodysplastic syndromes (MDS). Apart from intrinsic cellular mechanisms, various extrinsic factors of the BM immune microenvironment (IME) emerge as potential determinants of disease initiation and evolution. In MDS, the IME is reprogrammed, initially to prevent the development, but ultimately to support and provide a survival advantage to the dysplastic clone. Specific cellular elements, such as myeloid-derived suppressor cells (MDSCs) are recruited to support and enhance clonal expansion. The immune-mediated inhibition of normal hematopoiesis contributes to peripheral cytopenias of MDS patients, while immunosuppression in late-stage MDS enables immune evasion and disease progression towards acute myeloid leukemia (AML). In this review, we aim to elucidate the role of the mediators of immune response in the initial pathogenesis of MDS and the evolution of the disease. Full article
(This article belongs to the Special Issue Cancers Precision Immunotherapy)
Show Figures

Figure 1

31 pages, 1537 KiB  
Review
The Immune Contexture of Liposarcoma and Its Clinical Implications
by Antonia Resag, Giulia Toffanin, Iva Benešová, Luise Müller, Vlatko Potkrajcic, Andrej Ozaniak, Robert Lischke, Jirina Bartunkova, Antonio Rosato, Korinna Jöhrens, Franziska Eckert, Zuzana Strizova and Marc Schmitz
Cancers 2022, 14(19), 4578; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14194578 - 21 Sep 2022
Cited by 7 | Viewed by 3782
Abstract
Liposarcomas (LPS) are the most frequent malignancies in the soft tissue sarcoma family and consist of five distinctive histological subtypes, termed well-differentiated LPS, dedifferentiated LPS (DDLPS), myxoid LPS (MLPS), pleomorphic LPS, and myxoid pleomorphic LPS. They display variations in genetic alterations, clinical behavior, [...] Read more.
Liposarcomas (LPS) are the most frequent malignancies in the soft tissue sarcoma family and consist of five distinctive histological subtypes, termed well-differentiated LPS, dedifferentiated LPS (DDLPS), myxoid LPS (MLPS), pleomorphic LPS, and myxoid pleomorphic LPS. They display variations in genetic alterations, clinical behavior, and prognostic course. While accumulating evidence implicates a crucial role of the tumor immune contexture in shaping the response to anticancer treatments, the immunological landscape of LPS is highly variable across different subtypes. Thus, DDLPS is characterized by a higher abundance of infiltrating T cells, yet the opposite was reported for MLPS. Interestingly, a recent study indicated that the frequency of pre-existing T cells in soft tissue sarcomas has a predictive value for immune checkpoint inhibitor (CPI) therapy. Additionally, B cells and tertiary lymphoid structures were identified as potential biomarkers for the clinical outcome of LPS patients and response to CPI therapy. Furthermore, it was demonstrated that macrophages, predominantly of M2 polarization, are frequently associated with poor prognosis. An improved understanding of the complex LPS immune contexture enables the design and refinement of novel immunotherapeutic approaches. Here, we summarize recent studies focusing on the clinicopathological, genetic, and immunological determinants of LPS. Full article
(This article belongs to the Special Issue Cancers Precision Immunotherapy)
Show Figures

Graphical abstract

Back to TopTop