ijms-logo

Journal Browser

Journal Browser

Immunological Responses to Cancer Therapy

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Oncology".

Deadline for manuscript submissions: closed (31 December 2021) | Viewed by 73241

Special Issue Editor


E-Mail
Guest Editor
Amsterdam UMC
Interests: Immunotherapy of Cancer; Antibodies; Tumor Draining Lymph Nodes; Tumor Microenvironment; Ablative Therapy; T cells

Special Issue Information

Dear Colleagues,

Immunotherapy for cancer treatment has been the focus of many studies over the last two decades. Immunotherapy is remarkably effective, but unfortunately, only in a minority of patients, and immunological side-effects are a major concern. Combining strategies of standard ablative treatment with immunotherapeutics could be a way forward. For instance, combining ablative radiotherapy with immune-activating checkpoint blocker therapy could enhance clinical outcome. But also improving the design of treatment strategies, in terms of timing or administration route can lead to increase of efficacy. Clinical trials aimed to study if neo-adjuvant administration compared to adjuvant administration of checkpoint blockers leads to improved treatment efficacy are hot topics. And manuscripts describing local administration (intratumoral, or peritumoral) of immune-activating agents, such as TLR-ligands, checkpoint blockers or oncolytic viruses, are showing promising results in both pre-clinical and clinical studies. Furthermore, the design of novel personalized vaccine strategies against neoantigens, is a promising development, which could easily be combined with other treatment modalities.

In order to design the most efficient strategy or choose the most potent combination of treatments, it is vital to understand the underlying mechanism, responsible for treatment efficacy. Cellular and molecular immunological processes should be studied, in order to choose elements that compliment and not hamper each other. Timing and kinetics of immunological responses should be observed, for the right strategy-design to be made. Novel research strategies are therefore required for a better description of the mechanisms of these approaches for cancer therapy.

The focus of this Special Issue is to consider mechanistic understanding of immunological processes of different forms of cancer therapy, either immunotherapy or standard ablative therapy, as monotherapy or in combination with each other. This knowledge will aid clinicians to make informed choices about treatment combinations or design strategies. We will consider pre-clinical, translational and clinical studies that contain biomolecular analyses into immunological mechanisms of cancer treatment.

Dr. Marieke F. Fransen
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. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. 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

  • Immune response
  • Cancer
  • Ablation
  • Antigen presentation
  • Lymph nodes
  • Kinetics

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Published Papers (15 papers)

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

Editorial

Jump to: Research, Review

3 pages, 170 KiB  
Editorial
Immunological Responses to Cancer Therapy
by Marieke F. Fransen
Int. J. Mol. Sci. 2022, 23(13), 6989; https://doi.org/10.3390/ijms23136989 - 23 Jun 2022
Viewed by 1188
Abstract
The use of immunotherapy for cancer has taken flight in the last two decades, from experimental therapy envisioned mainly by laboratory scientists to everyday treatment used by physicians to treat many patients [...] Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)

Research

Jump to: Editorial, Review

19 pages, 1526 KiB  
Article
Deregulated Expression of Immune Checkpoints on Circulating CD4 T Cells May Complicate Clinical Outcome and Response to Treatment with Checkpoint Inhibitors in Multiple Myeloma Patients
by Anna Kulikowska de Nałęcz, Lidia Ciszak, Lidia Usnarska-Zubkiewicz, Irena Frydecka, Edyta Pawlak, Magdalena Szmyrka and Agata Kosmaczewska
Int. J. Mol. Sci. 2021, 22(17), 9298; https://doi.org/10.3390/ijms22179298 - 27 Aug 2021
Cited by 8 | Viewed by 2261
Abstract
Unlike solid-tumor patients, a disappointingly small subset of multiple myeloma (MM) patients treated with checkpoint inhibitors derive clinical benefits, suggesting differential participation of inhibitory receptors involved in the development of T-cell-mediated immunosuppression. In fact, T cells in MM patients have recently been shown [...] Read more.
Unlike solid-tumor patients, a disappointingly small subset of multiple myeloma (MM) patients treated with checkpoint inhibitors derive clinical benefits, suggesting differential participation of inhibitory receptors involved in the development of T-cell-mediated immunosuppression. In fact, T cells in MM patients have recently been shown to display features of immunosenescence and exhaustion involved in immune response inhibition. Therefore, we aimed to identify the dominant inhibitory pathway in MM patients to achieve its effective control by therapeutic interventions. By flow cytometry, we examined peripheral blood (PB) CD4 T cell characteristics assigned to senescence or exhaustion, considering PD-1, CTLA-4, and BTLA checkpoint expression, as well as secretory effector function, i.e., capacity for IFN-γ and IL-17 secretion. Analyses were performed in a total of 40 active myeloma patients (newly diagnosed and treated) and 20 healthy controls. At the single-cell level, we found a loss of studied checkpoints’ expression on MM CD4 T cells (both effector (Teff) and regulatory (Treg) cells) primarily at diagnosis; the checkpoint deficit in MM relapse was not significant. Nonetheless, PD-1 was the only checkpoint distributed on an increased proportion of T cells in all MM patients irrespective of disease phase, and its expression on CD4 Teff cells correlated with adverse clinical courses. Among patients, the relative defect in secretory effector function of CD4 T cells was more pronounced at myeloma relapse (as seen in declined Th1/Treg and Th17/Treg cell rates). Although the contribution of PD-1 to MM clinical outcomes is suggestive, our study clearly indicated that the inappropriate expression of immune checkpoints (associated with dysfunctionality of CD4 T cells and disease clinical phase) might be responsible for the sub-optimal clinical response to therapeutic checkpoint inhibitors in MM. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

15 pages, 6009 KiB  
Article
MTL-CEBPA Combined with Immunotherapy or RFA Enhances Immunological Anti-Tumor Response in Preclinical Models
by Kai-Wen Huang, Choon Ping Tan, Vikash Reebye, Cheng Ean Chee, Dimitris Zacharoulis, Robert Habib, David C. Blakey, John J. Rossi, Nagy Habib and Mikael H. Sodergren
Int. J. Mol. Sci. 2021, 22(17), 9168; https://doi.org/10.3390/ijms22179168 - 25 Aug 2021
Cited by 13 | Viewed by 4081
Abstract
The transcription factor CEBPA is a master regulator of liver homeostasis, myeloid cell differentiation and is downregulated in several oncogenic diseases. MTL-CEBPA is a small activating RNA drug which upregulates gene expression of CEBPA for treatment of hepatocellular carcinoma (HCC). We investigate whether [...] Read more.
The transcription factor CEBPA is a master regulator of liver homeostasis, myeloid cell differentiation and is downregulated in several oncogenic diseases. MTL-CEBPA is a small activating RNA drug which upregulates gene expression of CEBPA for treatment of hepatocellular carcinoma (HCC). We investigate whether MTL-CEBPA has immune modulatory effects by combining MTL-CEBPA with an anti-PD-1 checkpoint inhibitor (CPI) and/or radiofrequency ablation (RFA) in two preclinical models. First, mice with two flanks of HCC tumors (BNL) were treated with combinations of RFA (right flank), anti-PD-1 or MTL-CEBPA. The reduction of the left flank tumors was most pronounced in the group treated with RFA+anti-PD1+MTL-CEBPA and 7/8 animals responded. This was the only group with a significant increase in CD8+ and CD49b+/CD45+ tumor infiltrating lymphocytes (TIL). Second, a combination of anti-PD-1+MTL-CEBPA was tested in a CT26 colon cancer model and this treatment significantly reduced tumor size, modulated the tumor immune microenvironment and increased TILs. These data suggest a clinical role for combination treatment with CPIs, RFA and MTL-CEBPA through synergistic priming of the immune tumor response, enabling RFA and CPIs to have a pronounced anti-tumor effect including activity in non-treated tumors in the case of RFA. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

12 pages, 2086 KiB  
Article
Dendritic Cells Pulsed with Cytokine-Adjuvanted Tumor Membrane Vesicles Inhibit Tumor Growth in HER2-Positive and Triple Negative Breast Cancer Models
by Luis E. Munoz, Lenore Monterroza, Ramireddy Bommireddy, Yalda Shafizadeh, Christopher D. Pack, Sampath Ramachandiran, Shaker J. C. Reddy and Periasamy Selvaraj
Int. J. Mol. Sci. 2021, 22(16), 8377; https://doi.org/10.3390/ijms22168377 - 4 Aug 2021
Cited by 2 | Viewed by 3098
Abstract
Dendritic cells (DCs) are the most effective antigen presenting cells for the development of T cell responses. The only FDA approved DC-based immunotherapy to date is Sipuleucel-T, which utilizes a fusion protein to stimulate DCs ex vivo with GM-CSF and simultaneously deliver the [...] Read more.
Dendritic cells (DCs) are the most effective antigen presenting cells for the development of T cell responses. The only FDA approved DC-based immunotherapy to date is Sipuleucel-T, which utilizes a fusion protein to stimulate DCs ex vivo with GM-CSF and simultaneously deliver the antigen PAP for prostate cancer. This approach is restricted by the breadth of immunity elicited to a single antigen, and to cancers that have a defined tumor associated antigen. Other multi-antigen approaches have been restricted by poor efficacy of vaccine adjuvants. We have developed a vaccine platform that consists of autologous DCs pulsed with cytokine-adjuvanted tumor membrane vesicles (TMVs) made from tumor tissue, that encapsulate the antigenic landscape of individual tumors. Here we test the efficacy of DCs pulsed with TMVs incorporated with glycolipid-anchored immunostimulatory molecules (GPI-ISMs) in HER2-positive and triple negative breast cancer murine models. Pulsing of DCs with TMVs containing GPI-ISMs results in superior uptake of vesicles, DC activation and cytokine production. Adaptive transfer of TMV-pulsed DCs to tumor bearing mice results in the inhibition of tumor growth, reduction in lung metastasis, and an increase in immune cell infiltration into the tumors. These observations suggest that DCs pulsed with TMVs containing GPI-GM-CSF and GPI-IL-12 can be further developed to be used as a personalized immunotherapy platform for cancer treatment. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

22 pages, 21354 KiB  
Article
A Mathematical Description of the Bone Marrow Dynamics during CAR T-Cell Therapy in B-Cell Childhood Acute Lymphoblastic Leukemia
by Álvaro Martínez-Rubio, Salvador Chulián, Cristina Blázquez Goñi, Manuel Ramírez Orellana, Antonio Pérez Martínez, Alfonso Navarro-Zapata, Cristina Ferreras, Victor M. Pérez-García and María Rosa
Int. J. Mol. Sci. 2021, 22(12), 6371; https://doi.org/10.3390/ijms22126371 - 14 Jun 2021
Cited by 8 | Viewed by 3893
Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy has demonstrated high rates of response in recurrent B-cell Acute Lymphoblastic Leukemia in children and young adults. Despite this success, a fraction of patients’ experience relapse after treatment. Relapse is often preceded by recovery of healthy B [...] Read more.
Chimeric Antigen Receptor (CAR) T-cell therapy has demonstrated high rates of response in recurrent B-cell Acute Lymphoblastic Leukemia in children and young adults. Despite this success, a fraction of patients’ experience relapse after treatment. Relapse is often preceded by recovery of healthy B cells, which suggests loss or dysfunction of CAR T-cells in bone marrow. This site is harder to access, and thus is not monitored as frequently as peripheral blood. Understanding the interplay between B cells, leukemic cells, and CAR T-cells in bone marrow is paramount in ascertaining the causes of lack of response. In this paper, we put forward a mathematical model representing the interaction between constantly renewing B cells, CAR T-cells, and leukemic cells in the bone marrow. Our model accounts for the maturation dynamics of B cells and incorporates effector and memory CAR T-cells. The model provides a plausible description of the dynamics of the various cellular compartments in bone marrow after CAR T infusion. After exploration of the parameter space, we found that the dynamics of CAR T product and disease were independent of the dose injected, initial B-cell load, and leukemia burden. We also show theoretically the importance of CAR T product attributes in determining therapy outcome, and have studied a variety of possible response scenarios, including second dosage schemes. We conclude by setting out ideas for the refinement of the model. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

Review

Jump to: Editorial, Research

24 pages, 721 KiB  
Review
A Systematic Review of Expression and Immunogenicity of Human Endogenous Retroviral Proteins in Cancer and Discussion of Therapeutic Approaches
by Mikkel Dons Müller, Peter Johannes Holst and Karen Nørgaard Nielsen
Int. J. Mol. Sci. 2022, 23(3), 1330; https://doi.org/10.3390/ijms23031330 - 25 Jan 2022
Cited by 13 | Viewed by 5326
Abstract
Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections that have become fixed in the human genome. While HERV genes are typically silenced in healthy somatic cells, there are numerous reports of HERV transcription and translation across a wide spectrum of cancers, [...] Read more.
Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections that have become fixed in the human genome. While HERV genes are typically silenced in healthy somatic cells, there are numerous reports of HERV transcription and translation across a wide spectrum of cancers, while T and B cell responses against HERV proteins have been detected in cancer patients. This review systematically categorizes the published evidence on the expression of and adaptive immune response against specific HERVs in distinct cancer types. A systematic literature search was performed using Medical Search Headings (MeSH) in the PubMed/Medline database. Papers were included if they described the translational activity of HERVs. We present multiple tables that pair the protein expression of specific HERVs and cancer types with information on the quality of the evidence. We find that HERV-K is the most investigated HERV. HERV-W (syncytin-1) is the second-most investigated, while other HERVs have received less attention. From a therapeutic perspective, HERV-K and HERV-E are the only HERVs with experimental demonstration of effective targeted therapies, but unspecific approaches using antiviral and demethylating agents in combination with chemo- and immunotherapies have also been investigated. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

27 pages, 1803 KiB  
Review
Improving CAR T-Cell Persistence
by Violena Pietrobon, Lauren Anne Todd, Anghsumala Goswami, Ofir Stefanson, Zhifen Yang and Francesco Marincola
Int. J. Mol. Sci. 2021, 22(19), 10828; https://doi.org/10.3390/ijms221910828 - 7 Oct 2021
Cited by 50 | Viewed by 9234
Abstract
Over the last decade remarkable progress has been made in enhancing the efficacy of CAR T therapies. However, the clinical benefits are still limited, especially in solid tumors. Even in hematological settings, patients that respond to CAR T therapies remain at risk of [...] Read more.
Over the last decade remarkable progress has been made in enhancing the efficacy of CAR T therapies. However, the clinical benefits are still limited, especially in solid tumors. Even in hematological settings, patients that respond to CAR T therapies remain at risk of relapsing due to several factors including poor T-cell expansion and lack of long-term persistence after adoptive transfer. This issue is even more evident in solid tumors, as the tumor microenvironment negatively influences the survival, infiltration, and activity of T-cells. Limited persistence remains a significant hindrance to the development of effective CAR T therapies due to several determinants, which are encountered from the cell manufacturing step and onwards. CAR design and ex vivo manipulation, including culture conditions, may play a pivotal role. Moreover, previous chemotherapy and lymphodepleting treatments may play a relevant role. In this review, the main causes for decreased persistence of CAR T-cells in patients will be discussed, focusing on the molecular mechanisms underlying T-cell exhaustion. The approaches taken so far to overcome these limitations and to create exhaustion-resistant T-cells will be described. We will also examine the knowledge gained from several key clinical trials and highlight the molecular mechanisms determining T-cell stemness, as promoting stemness may represent an attractive approach to improve T-cell therapies. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Graphical abstract

28 pages, 2364 KiB  
Review
Ubiquitination in T-Cell Activation and Checkpoint Inhibition: New Avenues for Targeted Cancer Immunotherapy
by Shubhangi Gavali, Jianing Liu, Xinyi Li and Magdalena Paolino
Int. J. Mol. Sci. 2021, 22(19), 10800; https://doi.org/10.3390/ijms221910800 - 6 Oct 2021
Cited by 23 | Viewed by 7678
Abstract
The advent of T-cell-based immunotherapy has remarkably transformed cancer patient treatment. Despite their success, the currently approved immunotherapeutic protocols still encounter limitations, cause toxicity, and give disparate patient outcomes. Thus, a deeper understanding of the molecular mechanisms of T-cell activation and inhibition is [...] Read more.
The advent of T-cell-based immunotherapy has remarkably transformed cancer patient treatment. Despite their success, the currently approved immunotherapeutic protocols still encounter limitations, cause toxicity, and give disparate patient outcomes. Thus, a deeper understanding of the molecular mechanisms of T-cell activation and inhibition is much needed to rationally expand targets and possibilities to improve immunotherapies. Protein ubiquitination downstream of immune signaling pathways is essential to fine-tune virtually all immune responses, in particular, the positive and negative regulation of T-cell activation. Numerous studies have demonstrated that deregulation of ubiquitin-dependent pathways can significantly alter T-cell activation and enhance antitumor responses. Consequently, researchers in academia and industry are actively developing technologies to selectively exploit ubiquitin-related enzymes for cancer therapeutics. In this review, we discuss the molecular and functional roles of ubiquitination in key T-cell activation and checkpoint inhibitory pathways to highlight the vast possibilities that targeting ubiquitination offers for advancing T-cell-based immunotherapies. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

24 pages, 2242 KiB  
Review
Exploiting B Cell Transfer for Cancer Therapy: Engineered B Cells to Eradicate Tumors
by Audrey Page, Julie Hubert, Floriane Fusil and François-Loïc Cosset
Int. J. Mol. Sci. 2021, 22(18), 9991; https://doi.org/10.3390/ijms22189991 - 16 Sep 2021
Cited by 13 | Viewed by 9325
Abstract
Nowadays, cancers still represent a significant health burden, accounting for around 10 million deaths per year, due to ageing populations and inefficient treatments for some refractory cancers. Immunotherapy strategies that modulate the patient’s immune system have emerged as good treatment options. Among them, [...] Read more.
Nowadays, cancers still represent a significant health burden, accounting for around 10 million deaths per year, due to ageing populations and inefficient treatments for some refractory cancers. Immunotherapy strategies that modulate the patient’s immune system have emerged as good treatment options. Among them, the adoptive transfer of B cells selected ex vivo showed promising results, with a reduction in tumor growth in several cancer mouse models, often associated with antitumoral immune responses. Aside from the benefits of their intrinsic properties, including antigen presentation, antibody secretion, homing and long-term persistence, B cells can be modified prior to reinfusion to increase their therapeutic role. For instance, B cells have been modified mainly to boost their immuno-stimulatory activation potential by forcing the expression of costimulatory ligands using defined culture conditions or gene insertion. Moreover, tumor-specific antigen presentation by infused B cells has been increased by ex vivo antigen loading (peptides, RNA, DNA, virus) or by the sorting/ engineering of B cells with a B cell receptor specific to tumor antigens. Editing of the BCR also rewires B cell specificity toward tumor antigens, and may trigger, upon antigen recognition, the secretion of antitumor antibodies by differentiated plasma cells that can then be recognized by other immune components or cells involved in tumor clearance by antibody-dependent cell cytotoxicity or complement-dependent cytotoxicity for example. With the expansion of gene editing methodologies, new strategies to reprogram immune cells with whole synthetic circuits are being explored: modified B cells can sense disease-specific biomarkers and, in response, trigger the expression of therapeutic molecules, such as molecules that counteract the tumoral immunosuppressive microenvironment. Such strategies remain in their infancy for implementation in B cells, but are likely to expand in the coming years. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

19 pages, 1471 KiB  
Review
The Role of Somatic Mutations on the Immune Response of the Tumor Microenvironment in Prostate Cancer
by Camila Morais Melo, Thiago Vidotto, Luiz Paulo Chaves, William Lautert-Dutra, Rodolfo Borges dos Reis and Jeremy Andrew Squire
Int. J. Mol. Sci. 2021, 22(17), 9550; https://doi.org/10.3390/ijms22179550 - 2 Sep 2021
Cited by 18 | Viewed by 4787
Abstract
Immunotherapy has improved patient survival in many types of cancer, but for prostate cancer, initial results with immunotherapy have been disappointing. Prostate cancer is considered an immunologically excluded or cold tumor, unable to generate an effective T-cell response against cancer cells. However, a [...] Read more.
Immunotherapy has improved patient survival in many types of cancer, but for prostate cancer, initial results with immunotherapy have been disappointing. Prostate cancer is considered an immunologically excluded or cold tumor, unable to generate an effective T-cell response against cancer cells. However, a small but significant percentage of patients do respond to immunotherapy, suggesting that some specific molecular subtypes of this tumor may have a better response to checkpoint inhibitors. Recent findings suggest that, in addition to their function as cancer genes, somatic mutations of PTEN, TP53, RB1, CDK12, and DNA repair, or specific activation of regulatory pathways, such as ETS or MYC, may also facilitate immune evasion of the host response against cancer. This review presents an update of recent discoveries about the role that the common somatic mutations can play in changing the tumor microenvironment and immune response against prostate cancer. We describe how detailed molecular genetic analyses of the tumor microenvironment of prostate cancer using mouse models and human tumors are providing new insights into the cell types and pathways mediating immune responses. These analyses are helping researchers to design drug combinations that are more likely to target the molecular and immunological pathways that underlie treatment failure. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

18 pages, 799 KiB  
Review
Peripheral Blood-Based Biomarkers for Immune Checkpoint Inhibitors
by Ho Jung An, Hong Jae Chon and Chan Kim
Int. J. Mol. Sci. 2021, 22(17), 9414; https://doi.org/10.3390/ijms22179414 - 30 Aug 2021
Cited by 47 | Viewed by 5336
Abstract
As cancer immunotherapy using immune checkpoint inhibitors (ICIs) is rapidly evolving in clinical practice, it is necessary to identify biomarkers that will allow the selection of cancer patients who will benefit most or least from ICIs and to longitudinally monitor patients’ immune responses [...] Read more.
As cancer immunotherapy using immune checkpoint inhibitors (ICIs) is rapidly evolving in clinical practice, it is necessary to identify biomarkers that will allow the selection of cancer patients who will benefit most or least from ICIs and to longitudinally monitor patients’ immune responses during treatment. Various peripheral blood-based immune biomarkers are being identified with recent advances in high-throughput multiplexed analytical technologies. The identification of these biomarkers, which can be easily detected in blood samples using non-invasive and repeatable methods, will contribute to overcoming the limitations of previously used tissue-based biomarkers. Here, we discuss the potential of circulating immune cells, soluble immune and inflammatory molecules, circulating tumor cells and DNA, exosomes, and the blood-based tumor mutational burden, as biomarkers for the prediction of immune responses and clinical benefit from ICI treatment in patients with advanced cancer. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

10 pages, 1178 KiB  
Review
Immune Checkpoint Therapy: Tumor Draining Lymph Nodes in the Spotlights
by Marieke F. Fransen, Thorbald van Hall and Ferry Ossendorp
Int. J. Mol. Sci. 2021, 22(17), 9401; https://doi.org/10.3390/ijms22179401 - 30 Aug 2021
Cited by 19 | Viewed by 4322
Abstract
Tumor-draining lymph nodes play a paradoxical role in cancer. Surgeons often resect these sentinel lymph nodes to determine metastatic spread, thereby enabling prognosis and treatment. However, lymph nodes are vital organs for the orchestration of immune responses, due to the close encounters of [...] Read more.
Tumor-draining lymph nodes play a paradoxical role in cancer. Surgeons often resect these sentinel lymph nodes to determine metastatic spread, thereby enabling prognosis and treatment. However, lymph nodes are vital organs for the orchestration of immune responses, due to the close encounters of dedicated immune cells. In view of the success of immunotherapy, the removal of tumor-draining lymph nodes needs to be re-evaluated and viewed in a different light. Recently, an important role for tumor-draining lymph nodes has been proposed in the immunotherapy of cancer. This new insight can change the use of immune checkpoint therapy, particularly with respect to the use in neoadjuvant settings in which lymph nodes are still operational. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

14 pages, 2772 KiB  
Review
Local Therapies and Modulation of Tumor Surrounding Stroma in Malignant Pleural Mesothelioma: A Translational Approach
by Daniela Lisini, Sara Lettieri, Sara Nava, Giulia Accordino, Simona Frigerio, Chandra Bortolotto, Andrea Lancia, Andrea Riccardo Filippi, Francesco Agustoni, Laura Pandolfi, Davide Piloni, Patrizia Comoli, Angelo Guido Corsico and Giulia Maria Stella
Int. J. Mol. Sci. 2021, 22(16), 9014; https://doi.org/10.3390/ijms22169014 - 20 Aug 2021
Cited by 6 | Viewed by 2843
Abstract
Malignant Pleural Mesothelioma (MPM) is a rare and aggressive neoplasm of the pleural mesothelium, mainly associated with asbestos exposure and still lacking effective therapies. Modern targeted biological strategies that have revolutionized the therapy of other solid tumors have not had success so far [...] Read more.
Malignant Pleural Mesothelioma (MPM) is a rare and aggressive neoplasm of the pleural mesothelium, mainly associated with asbestos exposure and still lacking effective therapies. Modern targeted biological strategies that have revolutionized the therapy of other solid tumors have not had success so far in the MPM. Combination immunotherapy might achieve better results over chemotherapy alone, but there is still a need for more effective therapeutic approaches. Based on the peculiar disease features of MPM, several strategies for local therapeutic delivery have been developed over the past years. The common rationale of these approaches is: (i) to reduce the risk of drug inactivation before reaching the target tumor cells; (ii) to increase the concentration of active drugs in the tumor micro-environment and their bioavailability; (iii) to reduce toxic effects on normal, non-transformed cells, because of much lower drug doses than those used for systemic chemotherapy. The complex interactions between drugs and the local immune-inflammatory micro-environment modulate the subsequent clinical response. In this perspective, the main interest is currently addressed to the development of local drug delivery platforms, both cell therapy and engineered nanotools. We here propose a review aimed at deep investigation of the biologic effects of the current local therapies for MPM, including cell therapies, and the mechanisms of interaction with the tumor micro-environment. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

30 pages, 760 KiB  
Review
T-Cell Responses in Merkel Cell Carcinoma: Implications for Improved Immune Checkpoint Blockade and Other Therapeutic Options
by Laura Gehrcken, Tatjana Sauerer, Niels Schaft and Jan Dörrie
Int. J. Mol. Sci. 2021, 22(16), 8679; https://doi.org/10.3390/ijms22168679 - 12 Aug 2021
Cited by 6 | Viewed by 3844
Abstract
Merkel cell carcinoma (MCC) is a rare and aggressive skin cancer with rising incidence and high mortality. Approximately 80% of the cases are caused by the human Merkel cell polyomavirus, while the remaining 20% are induced by UV light leading to mutations. The [...] Read more.
Merkel cell carcinoma (MCC) is a rare and aggressive skin cancer with rising incidence and high mortality. Approximately 80% of the cases are caused by the human Merkel cell polyomavirus, while the remaining 20% are induced by UV light leading to mutations. The standard treatment of metastatic MCC is the use of anti-PD-1/-PD-L1-immune checkpoint inhibitors (ICI) such as Pembrolizumab or Avelumab, which in comparison with conventional chemotherapy show better overall response rates and longer duration of responses in patients. Nevertheless, 50% of the patients do not respond or develop ICI-induced, immune-related adverse events (irAEs), due to diverse mechanisms, such as down-regulation of MHC complexes or the induction of anti-inflammatory cytokines. Other immunotherapeutic options such as cytokines and pro-inflammatory agents or the use of therapeutic vaccination offer great ameliorations to ICI. Cytotoxic T-cells play a major role in the effectiveness of ICI, and tumour-infiltrating CD8+ T-cells and their phenotype contribute to the clinical outcome. This literature review presents a summary of current and future checkpoint inhibitor therapies in MCC and demonstrates alternative therapeutic options. Moreover, the importance of T-cell responses and their beneficial role in MCC treatment is discussed. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

22 pages, 1453 KiB  
Review
Autoimmune Responses in Oncology: Causes and Significance
by Halin Bareke, Pablo Juanes-Velasco, Alicia Landeira-Viñuela, Angela-Patricia Hernandez, Juan Jesús Cruz, Lorena Bellido, Emilio Fonseca, Alfonssina Niebla-Cárdenas, Enrique Montalvillo, Rafael Góngora and Manuel Fuentes
Int. J. Mol. Sci. 2021, 22(15), 8030; https://doi.org/10.3390/ijms22158030 - 27 Jul 2021
Cited by 12 | Viewed by 4266
Abstract
Specific anti-tumor immune responses have proven to be pivotal in shaping tumorigenesis and tumor progression in solid cancers. These responses can also be of an autoimmune nature, and autoantibodies can sometimes be present even before the onset of clinically overt disease. Autoantibodies can [...] Read more.
Specific anti-tumor immune responses have proven to be pivotal in shaping tumorigenesis and tumor progression in solid cancers. These responses can also be of an autoimmune nature, and autoantibodies can sometimes be present even before the onset of clinically overt disease. Autoantibodies can be generated due to mutated gene products, aberrant expression and post-transcriptional modification of proteins, a pro-immunogenic milieu, anti-cancer treatments, cross-reactivity of tumor-specific lymphocytes, epitope spreading, and microbiota-related and genetic factors. Understanding these responses has implications for both basic and clinical immunology. Autoantibodies in solid cancers can be used for early detection of cancer as well as for biomarkers of prognosis and treatment response. High-throughput techniques such as protein microarrays make parallel detection of multiple autoantibodies for increased specificity and sensitivity feasible, affordable, and quick. Cancer immunotherapy has revolutionized cancer treatments and has made a considerable impact on reducing cancer-associated morbidity and mortality. However, immunotherapeutic interventions such as immune checkpoint inhibition can induce immune-related toxicities, which can even be life-threatening. Uncovering the reasons for treatment-induced autoimmunity can lead to fine-tuning of cancer immunotherapy approaches to evade toxic events while inducing an effective anti-tumor immune response. Full article
(This article belongs to the Special Issue Immunological Responses to Cancer Therapy)
Show Figures

Figure 1

Back to TopTop