Immuno-Oncology: Identification of Therapeutic Targets, Development of Novel Drugs and Improvement of Treatment Approaches for Cancer

A special issue of Journal of Clinical Medicine (ISSN 2077-0383). This special issue belongs to the section "Pharmacology".

Deadline for manuscript submissions: closed (31 July 2020) | Viewed by 32741

Special Issue Editor


E-Mail
Guest Editor
Research Scientist, Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
Interests: cancer genetics; small noncoding RNAs dysregulation in cancer; gene expression dysregulation in cancer; targeted therapy for cancer; chronic lymphocytic leukemia; lung cancer

Special Issue Information

Dear Colleagues

Recently, we witnessed an extraordinary development in cancer therapy, led by the evolving field of immuno–oncology. Traditional chemotherapy destroys cancer cells, but it also affects healthy cells. To overcome this dilemma, researchers have tried several approaches. Hormonal therapies inhibit growth-promoting hormones or block their survival signal to cancer cells, but this strategy only works for certain cancer types derived from hormonally responsive tissues. Thus, compounds targeting cancer-specific mutated genes were generated, and more target molecules are currently being investigated to develop new drugs. Immunotherapy is an evolution of targeted therapy where the immune system is targeted to improve its ability to kill cancer cells. Passive immunotherapies enhance the immune system’s antitumor response using monoclonal antibodies, and the aim of current research is to optimize this strategy by developing personalized treatments. Active immunotherapy directly stimulates the immune system to kill cancer cells either by targeting immune-checkpoints inhibitors (thus interfering with the tumor’s ability to inhibit the immune system from attacking cancer cells) or by engineering patient T cells to recognize and kill cancer cells more efficiently (CAR-T therapy). Additionally, effective vaccines are already employed for infectious agent-related cancers and immunoprevention of cancers not related to infectious agents was observed in transgenic mice with activated oncogenes, indicating that stimulation of the immune system in healthy hosts can inhibit carcinogenesis. These strategies, are having an extraordinary success, but they can have side effects. Since these approaches are quickly taking over classic chemotherapy, perfecting immuno–oncology is essential to improve patient wellbeing. Hence, in this issue, we will discuss the most recent discoveries leading to the development of a new generation of drugs and therapeutic approaches for cancer treatment.

Dr. Veronica Balatti
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. Journal of Clinical Medicine 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 2600 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

  • Cancer treatments
  • Molecular genetics of cancer
  • Targeted therapy
  • Immunotherapy
  • Monoclonal antibody therapy
  • Cytokine therapy
  • Checkpoint inhibitors
  • CAR-T cells
  • Combination therapy
  • Cancer immunoprevention

Published Papers (6 papers)

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

Review

Jump to: Other

25 pages, 647 KiB  
Review
A Holistic Perspective: Exosomes Shuttle between Nerves and Immune Cells in the Tumor Microenvironment
by Mihnea P. Dragomir, Vlad Moisoiu, Roxana Manaila, Barbara Pardini, Erik Knutsen, Simone Anfossi, Moran Amit and George A. Calin
J. Clin. Med. 2020, 9(11), 3529; https://doi.org/10.3390/jcm9113529 - 31 Oct 2020
Cited by 9 | Viewed by 2960
Abstract
One of the limitations of cancer research has been the restricted focus on tumor cells and the omission of other non-malignant cells that are constitutive elements of this systemic disease. Current research is focused on the bidirectional communication between tumor cells and other [...] Read more.
One of the limitations of cancer research has been the restricted focus on tumor cells and the omission of other non-malignant cells that are constitutive elements of this systemic disease. Current research is focused on the bidirectional communication between tumor cells and other components of the tumor microenvironment (TME), such as immune and endothelial cells, and nerves. A major success of this bidirectional approach has been the development of immunotherapy. Recently, a more complex landscape involving a multi-lateral communication between the non-malignant components of the TME started to emerge. A prime example is the interplay between immune and endothelial cells, which led to the approval of anti-vascular endothelial growth factor-therapy combined with immune checkpoint inhibitors and classical chemotherapy in non-small cell lung cancer. Hence, a paradigm shift approach is to characterize the crosstalk between different non-malignant components of the TME and understand their role in tumorigenesis. In this perspective, we discuss the interplay between nerves and immune cells within the TME. In particular, we focus on exosomes and microRNAs as a systemic, rapid and dynamic communication channel between tumor cells, nerves and immune cells contributing to cancer progression. Finally, we discuss how combinatorial therapies blocking this tumorigenic cross-talk could lead to improved outcomes for cancer patients. Full article
Show Figures

Figure 1

57 pages, 1376 KiB  
Review
Epithelial Ovarian Cancer and the Immune System: Biology, Interactions, Challenges and Potential Advances for Immunotherapy
by Anne M. Macpherson, Simon C. Barry, Carmela Ricciardelli and Martin K. Oehler
J. Clin. Med. 2020, 9(9), 2967; https://doi.org/10.3390/jcm9092967 - 14 Sep 2020
Cited by 21 | Viewed by 4362
Abstract
Recent advances in the understanding of immune function and the interactions with tumour cells have led to the development of various cancer immunotherapies and strategies for specific cancer types. However, despite some stunning successes with some malignancies such as melanomas and lung cancer, [...] Read more.
Recent advances in the understanding of immune function and the interactions with tumour cells have led to the development of various cancer immunotherapies and strategies for specific cancer types. However, despite some stunning successes with some malignancies such as melanomas and lung cancer, most patients receive little or no benefit from immunotherapy, which has been attributed to the tumour microenvironment and immune evasion. Although the US Food and Drug Administration have approved immunotherapies for some cancers, to date, only the anti-angiogenic antibody bevacizumab is approved for the treatment of epithelial ovarian cancer. Immunotherapeutic strategies for ovarian cancer are still under development and being tested in numerous clinical trials. A detailed understanding of the interactions between cancer and the immune system is vital for optimisation of immunotherapies either alone or when combined with chemotherapy and other therapies. This article, in two main parts, provides an overview of: (1) components of the normal immune system and current knowledge regarding tumour immunology, biology and their interactions; (2) strategies, and targets, together with challenges and potential innovative approaches for cancer immunotherapy, with attention given to epithelial ovarian cancer. Full article
Show Figures

Figure 1

29 pages, 944 KiB  
Review
Role of Non-Coding RNAs in the Development of Targeted Therapy and Immunotherapy Approaches for Chronic Lymphocytic Leukemia
by Felice Pepe and Veronica Balatti
J. Clin. Med. 2020, 9(2), 593; https://doi.org/10.3390/jcm9020593 - 21 Feb 2020
Cited by 13 | Viewed by 4445
Abstract
In the past decade, novel targeted therapy approaches, such as BTK inhibitors and Bcl2 blockers, and innovative treatments that regulate the immune response against cancer cells, such as monoclonal antibodies, CAR-T cell therapy, and immunomodulatory molecules, have been established to provide support for [...] Read more.
In the past decade, novel targeted therapy approaches, such as BTK inhibitors and Bcl2 blockers, and innovative treatments that regulate the immune response against cancer cells, such as monoclonal antibodies, CAR-T cell therapy, and immunomodulatory molecules, have been established to provide support for the treatment of patients. However, drug resistance development and relapse are still major challenges in CLL treatment. Several studies revealed that non-coding RNAs have a main role in the development and progression of CLL. Specifically, microRNAs (miRs) and tRNA-derived small-RNAs (tsRNAs) were shown to be outstanding biomarkers that can be used to diagnose and monitor the disease and to possibly anticipate drug resistance and relapse, thus supporting physicians in the selection of treatment regimens tailored to the patient needs. In this review, we will summarize the most recent discoveries in the field of targeted therapy and immunotherapy for CLL and discuss the role of ncRNAs in the development of novel drugs and combination regimens for CLL patients. Full article
Show Figures

Figure 1

19 pages, 834 KiB  
Review
Chimeric Antigen Receptor T-Cell Therapy for Colorectal Cancer
by Daniel Sur, Andrei Havasi, Calin Cainap, Gabriel Samasca, Claudia Burz, Ovidiu Balacescu, Iulia Lupan, Diana Deleanu and Alexandru Irimie
J. Clin. Med. 2020, 9(1), 182; https://doi.org/10.3390/jcm9010182 - 9 Jan 2020
Cited by 45 | Viewed by 11525
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a new genetically engineered method of immunotherapy for cancer. The patient’s T-cells are modified to express a specific receptor that sticks to the tumor antigen. This modified cell is then reintroduced into the patient’s body to [...] Read more.
Chimeric antigen receptor (CAR) T-cell therapy represents a new genetically engineered method of immunotherapy for cancer. The patient’s T-cells are modified to express a specific receptor that sticks to the tumor antigen. This modified cell is then reintroduced into the patient’s body to fight the resilient cancer cells. After exhibiting positive results in hematological malignancies, this therapy is being proposed for solid tumors like colorectal cancer. The clinical data of CAR T-cell therapy in colorectal cancer is rather scarce. In this review, we summarize the current state of knowledge, challenges, and future perspectives of CAR T-cell therapy in colorectal cancer. A total of 22 articles were included in this review. Eligible studies were selected and reviewed by two researchers from 49 articles found on Pubmed, Web of Science, and clinicaltrials.gov. This therapy, at the moment, provides modest benefits in solid tumors. Not taking into consideration the high manufacturing and retail prices, there are still limitations like increased toxicities, relapses, and unfavorable tumor microenvironment for CAR T-cell therapy in colorectal cancer. Full article
Show Figures

Figure 1

20 pages, 1392 KiB  
Review
Targeting of the Cancer-Associated Fibroblast—T-Cell Axis in Solid Malignancies
by Tom J. Harryvan, Els M. E. Verdegaal, James C. H. Hardwick, Lukas J. A. C. Hawinkels and Sjoerd H. van der Burg
J. Clin. Med. 2019, 8(11), 1989; https://doi.org/10.3390/jcm8111989 - 15 Nov 2019
Cited by 45 | Viewed by 6088
Abstract
The introduction of a wide range of immunotherapies in clinical practice has revolutionized the treatment of cancer in the last decade. The majority of these therapeutic modalities are centered on reinvigorating a tumor-reactive cytotoxic T-cell response. While impressive clinical successes are obtained, the [...] Read more.
The introduction of a wide range of immunotherapies in clinical practice has revolutionized the treatment of cancer in the last decade. The majority of these therapeutic modalities are centered on reinvigorating a tumor-reactive cytotoxic T-cell response. While impressive clinical successes are obtained, the majority of cancer patients still fail to show a clinical response, despite the fact that their tumors express antigens that can be recognized by the immune system. This is due to a series of other cellular actors, present in or attracted towards the tumor microenvironment, including regulatory T-cells, myeloid-derived suppressor cells and cancer-associated fibroblasts (CAFs). As the main cellular constituent of the tumor-associated stroma, CAFs form a heterogeneous group of cells which can drive cancer cell invasion but can also impair the migration and activation of T-cells through direct and indirect mechanisms. This singles CAFs out as an important next target for further optimization of T-cell based immunotherapies. Here, we review the recent literature on the role of CAFs in orchestrating T-cell activation and migration within the tumor microenvironment and discuss potential avenues for targeting the interactions between fibroblasts and T-cells. Full article
Show Figures

Figure 1

Other

Jump to: Review

11 pages, 911 KiB  
Brief Report
The Antitumor Effect of Heparin is not Mediated by Direct NK Cell Activation
by Gustavo R. Rossi, Jenifer P. Gonçalves, Timothy McCulloch, Rebecca B. Delconte, Robert J. Hennessy, Nicholas D. Huntington, Edvaldo S. Trindade and Fernando Souza-Fonseca-Guimaraes
J. Clin. Med. 2020, 9(8), 2666; https://doi.org/10.3390/jcm9082666 - 18 Aug 2020
Cited by 7 | Viewed by 2825
Abstract
Natural killer (NK) cells are innate lymphocytes responsible for the elimination of infected or transformed cells. The activation or inhibition of NK cells is determined by the balance of target cell ligand recognition by stimulatory and inhibitory receptors on their surface. Previous reports [...] Read more.
Natural killer (NK) cells are innate lymphocytes responsible for the elimination of infected or transformed cells. The activation or inhibition of NK cells is determined by the balance of target cell ligand recognition by stimulatory and inhibitory receptors on their surface. Previous reports have suggested that the glycosaminoglycan heparin is a ligand for the natural cytotoxicity receptors NKp30, NKp44 (human), and NKp46 (both human and mouse). However, the effects of heparin on NK cell homeostasis and function remain unclear. Here, we show that heparin does not enhance NK cell proliferation or killing through NK cell activation. Alternatively, in mice models, heparin promoted NK cell survival in vitro and controlled B16-F10 melanoma metastasis development in vivo. In human NK cells, heparin promisingly increased interferon (IFN)-γ production in synergy with IL-12, although the mechanism remains elusive. Our data showed that heparin is not able to increase NK cell cytotoxicity. Full article
Show Figures

Figure 1

Back to TopTop