ijms-logo

Journal Browser

Journal Browser

Dendritic Cell and Cancer Therapy

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

Deadline for manuscript submissions: closed (31 May 2022) | Viewed by 26399

Special Issue Editor


E-Mail
Guest Editor
Haematology-Oncology and Stem Cell Transplantation Unit, Department of Haematology and Developmental Therapeutics, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italia
Interests: dendritic cells; immunotherapy; gamma-delta T cells; cancer biomarkers; tumor immunology; cytometry
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The strategies for cancer immunotherapy have obtained mixed results mainly due to immune suppression and several immune escape mechanisms induced by tumor cells. In this regard, Dendritic Cells have become a hopeful instrument for cancer vaccines that aims to re-educate the immune system, leading to a potent anti-cancer response that is able to overcome the immunosuppressive tumor microenvironment. However, cancer cells can deregulate Dendritic Cell function by a combination of different mechanisms, including antigen down-regulation and secretion of molecules that together adjust the immune-stimulating or the immune-suppressive functional plasticity of Dendritic Cells. 

Therefore, new insights will be helpful to define molecules and pathways that can modulate the performance of Dendritic Cell subsets. In particular, a better comprehension of the complex interactions occurring in the tumor microenvironment will undoubtedly improve the design of really efficient and durable immunotherapeutic approaches for tumor disease control.

Dr. Domenico Galati
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

  • dendritic cells
  • dendritic cell subsets
  • cancer immunotherapy
  • immune suppression
  • cancer vaccines
  • anti-cancer response
  • tumor microenvironment

Related Special Issue

Published Papers (8 papers)

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

Editorial

Jump to: Research, Review

3 pages, 169 KiB  
Editorial
Dendritic Cell and Cancer Therapy
by Domenico Galati and Serena Zanotta
Int. J. Mol. Sci. 2023, 24(4), 4253; https://doi.org/10.3390/ijms24044253 - 20 Feb 2023
Cited by 3 | Viewed by 2719
Abstract
Dendritic cells (DCs) are acknowledged as the most potent professional antigen-presenting cells (APCs), able to induce adaptive immunity and support the innate immune response [...] Full article
(This article belongs to the Special Issue Dendritic Cell and Cancer Therapy)

Research

Jump to: Editorial, Review

23 pages, 2460 KiB  
Article
Dendritic Cell-Triggered Immune Activation Goes along with Provision of (Leukemia-Specific) Integrin Beta 7-Expressing Immune Cells and Improved Antileukemic Processes
by Elias Rackl, Lin Li, Lara Kristina Klauer, Selda Ugur, Elena Pepeldjiyska, Corinna L. Seidel, Carina Gunsilius, Melanie Weinmann, Fatemeh Doraneh-Gard, Nina Reiter, Caroline Plett, Daniel Christoph Amberger, Peter Bojko, Doris Kraemer, Jörg Schmohl, Andreas Rank, Christoph Schmid and Helga Maria Schmetzer
Int. J. Mol. Sci. 2023, 24(1), 463; https://doi.org/10.3390/ijms24010463 - 27 Dec 2022
Cited by 4 | Viewed by 1901
Abstract
Integrin beta 7 (β7), a subunit of the integrin receptor, is expressed on the surface of immune cells and mediates cell–cell adhesions and interactions, e.g., antitumor or autoimmune reactions. Here, we analyzed, whether the stimulation of immune cells by dendritic cells (of leukemic [...] Read more.
Integrin beta 7 (β7), a subunit of the integrin receptor, is expressed on the surface of immune cells and mediates cell–cell adhesions and interactions, e.g., antitumor or autoimmune reactions. Here, we analyzed, whether the stimulation of immune cells by dendritic cells (of leukemic derivation in AML patients or of monocyte derivation in healthy donors) leads to increased/leukemia-specific β7 expression in immune cells after T-cell-enriched mixed lymphocyte culture—finally leading to improved antileukemic cytotoxicity. Healthy, as well as AML and MDS patients’ whole blood (WB) was treated with Kit-M (granulocyte–macrophage colony-stimulating factor (GM-CSF) + prostaglandin E1 (PGE1)) or Kit-I (GM-CSF + Picibanil) in order to generate DCs (DCleu or monocyte-derived DC), which were then used as stimulator cells in MLC. To quantify antigen/leukemia-specific/antileukemic functionality, a degranulation assay (DEG), an intracellular cytokine assay (INTCYT) and a cytotoxicity fluorolysis assay (CTX) were used. (Leukemia-specific) cell subtypes were quantified via flow cytometry. The Kit treatment of WB (compared to the control) resulted in the generation of DC/DCleu, which induced increased activation of innate and adaptive cells after MLC. Kit-pretreated WB (vs. the control) led to significantly increased frequencies of β7-expressing T-cells, degranulating and intracellular cytokine-producing β7-expressing immune cells and, in patients’ samples, increased blast lysis. Positive correlations were found between the Kit-M-mediated improvement of blast lysis (vs. the control) and frequencies of β7-expressing T-cells. Our findings indicate that DC-based immune therapies might be able to specifically activate the immune system against blasts going along with increased frequencies of (leukemia-specific) β7-expressing immune cells. Furthermore, β7 might qualify as a predictor for the efficiency and the success of AML and/or MDS therapies. Full article
(This article belongs to the Special Issue Dendritic Cell and Cancer Therapy)
Show Figures

Figure 1

13 pages, 27547 KiB  
Article
Immunostimulatory Potential of Extracellular Vesicles Isolated from an Edible Plant, Petasites japonicus, via the Induction of Murine Dendritic Cell Maturation
by Jeong Moo Han, Ha-Yeon Song, Seung-Taik Lim, Kwang-Il Kim, Ho Seong Seo and Eui-Baek Byun
Int. J. Mol. Sci. 2021, 22(19), 10634; https://doi.org/10.3390/ijms221910634 - 30 Sep 2021
Cited by 22 | Viewed by 2267
Abstract
Extracellular vesicles (EVs) have recently been isolated from different plants. Plant-derived EVs have been proposed as potent therapeutics and drug-delivery nanoplatforms for delivering biomolecules, including proteins, RNAs, DNAs, and lipids. Herein, Petasites japonicus-derived EVs (PJ-EVs) were isolated through a series of centrifugation [...] Read more.
Extracellular vesicles (EVs) have recently been isolated from different plants. Plant-derived EVs have been proposed as potent therapeutics and drug-delivery nanoplatforms for delivering biomolecules, including proteins, RNAs, DNAs, and lipids. Herein, Petasites japonicus-derived EVs (PJ-EVs) were isolated through a series of centrifugation steps and characterized using dynamic light scattering and transmission electron microscopy. Immunomodulatory effects of PJ-EVs were assessed using dendritic cells (DCs). PJ-EVs exhibited a spherical morphology with an average size of 122.6 nm. They induced the maturation of DCs via an increase in the expression of surface molecules (CD80, CD86, MHC-I, and MHC-II), production of Th1-polarizing cytokines (TNF-α and IL-12p70), and antigen-presenting ability; however, they reduced the antigen-uptake ability. Furthermore, maturation of DCs induced by PJ-EVs was dependent on the activation and phosphorylation of MAPK and NF-κB signal pathways. Notably, PJ-EV-treated DCs strongly induced the proliferation and differentiation of naïve T cells toward Th1-type T cells and cytotoxic CD8+ T cells along with robust secretion of IFN-γ and IL-2. In conclusion, our study indicates that PJ-EVs can be potent immunostimulatory candidates with an ability of strongly inducing the maturation of DCs. Full article
(This article belongs to the Special Issue Dendritic Cell and Cancer Therapy)
Show Figures

Figure 1

15 pages, 22167 KiB  
Article
Intranasal Administration of Codium fragile Polysaccharide Elicits Anti-Cancer Immunity against Lewis Lung Carcinoma
by Yuhua Wang, Eun-Koung An, So-Jung Kim, SangGuan You and Jun-O Jin
Int. J. Mol. Sci. 2021, 22(19), 10608; https://doi.org/10.3390/ijms221910608 - 30 Sep 2021
Cited by 11 | Viewed by 2310
Abstract
Natural polysaccharides have shown promising effects on the regulation of immunity in animals. In this study, we examined the immune stimulatory effect of intranasally administered Codium fragile polysaccharides (CFPs) in mice. Intranasal administration of CFPs in C57BL/6 mice induced the upregulation of surface [...] Read more.
Natural polysaccharides have shown promising effects on the regulation of immunity in animals. In this study, we examined the immune stimulatory effect of intranasally administered Codium fragile polysaccharides (CFPs) in mice. Intranasal administration of CFPs in C57BL/6 mice induced the upregulation of surface activation marker expression in macrophages and dendritic cells (DCs) in the mediastinal lymph node (mLN) and the production of interleukin-6 (IL-6), IL-12p70, and tumor necrosis factor-α in bronchoalveolar lavage fluid. Moreover, the number of conventional DCs (cDCs) was increased in the mLNs by the upregulation of C-C motif chemokine receptor 7 expression, and subsets of cDCs were also activated following the intranasal administration of CFP. In addition, the intranasal administration of CFPs promoted the activation of natural killer (NK) and T cells in the mLNs, which produce pro-inflammatory cytokines and cytotoxic mediators. Finally, daily administration of CFPs inhibited the infiltration of Lewis lung carcinoma cells into the lungs, and the preventive effect of CFPs on tumor growth required NK and CD8 T cells. Furthermore, CFPs combined with anti-programmed cell death-ligand 1 (PD-L1) antibody (Ab) improved the therapeutic effect of anti-PD-L1 Ab against lung cancer. Therefore, these data demonstrated that the intranasal administration of CFP induced mucosal immunity against lung cancer. Full article
(This article belongs to the Special Issue Dendritic Cell and Cancer Therapy)
Show Figures

Figure 1

16 pages, 3433 KiB  
Article
Enhancement of Immune Checkpoint Inhibitor-Mediated Anti-Cancer Immunity by Intranasal Treatment of Ecklonia cava Fucoidan against Metastatic Lung Cancer
by Wei Zhang, Juyoung Hwang, Dhananjay Yadav, Eun-Koung An, Minseok Kwak, Peter Chang-Whan Lee and Jun-O Jin
Int. J. Mol. Sci. 2021, 22(17), 9125; https://doi.org/10.3390/ijms22179125 - 24 Aug 2021
Cited by 22 | Viewed by 2556
Abstract
Although fucoidan, a well-studied seaweed-extracted polysaccharide, has shown immune stimulatory effects that elicit anticancer immunity, mucosal adjuvant effects via intranasal administration have not been studied. In this study, the effect of Ecklonia cava-extracted fucoidan (ECF) on the induction of anti-cancer immunity in [...] Read more.
Although fucoidan, a well-studied seaweed-extracted polysaccharide, has shown immune stimulatory effects that elicit anticancer immunity, mucosal adjuvant effects via intranasal administration have not been studied. In this study, the effect of Ecklonia cava-extracted fucoidan (ECF) on the induction of anti-cancer immunity in the lung was examined by intranasal administration. In C57BL/6 and BALB/c mice, intranasal administration of ECF promoted the activation of dendritic cells (DCs), natural killer (NK) cells, and T cells in the mediastinal lymph node (mLN). The ECF-induced NK and T cell activation was mediated by DCs. In addition, intranasal injection with ECF enhanced the anti-PD-L1 antibody-mediated anti-cancer activities against B16 melanoma and CT-26 carcinoma tumor growth in the lungs, which were required cytotoxic T lymphocytes and NK cells. Thus, these data demonstrated that ECF functioned as a mucosal adjuvant that enhanced the immunotherapeutic effect of immune checkpoint inhibitors against metastatic lung cancer. Full article
(This article belongs to the Special Issue Dendritic Cell and Cancer Therapy)
Show Figures

Figure 1

Review

Jump to: Editorial, Research

24 pages, 1839 KiB  
Review
Dendritic Cell-Based Immunotherapy in Hot and Cold Tumors
by Byeong Hoon Kang and Heung Kyu Lee
Int. J. Mol. Sci. 2022, 23(13), 7325; https://doi.org/10.3390/ijms23137325 - 30 Jun 2022
Cited by 7 | Viewed by 4673
Abstract
Dendritic cells mediate innate and adaptive immune responses and are directly involved in the activation of cytotoxic T lymphocytes that kill tumor cells. Dendritic cell-based cancer immunotherapy has clinical benefits. Dendritic cell subsets are diverse, and tumors can be hot or cold, depending [...] Read more.
Dendritic cells mediate innate and adaptive immune responses and are directly involved in the activation of cytotoxic T lymphocytes that kill tumor cells. Dendritic cell-based cancer immunotherapy has clinical benefits. Dendritic cell subsets are diverse, and tumors can be hot or cold, depending on their immunogenicity; this heterogeneity affects the success of dendritic cell-based immunotherapy. Here, we review the ontogeny of dendritic cells and dendritic cell subsets. We also review the characteristics of hot and cold tumors and briefly introduce therapeutic trials related to hot and cold tumors. Lastly, we discuss dendritic cell-based cancer immunotherapy in hot and cold tumors. Full article
(This article belongs to the Special Issue Dendritic Cell and Cancer Therapy)
Show Figures

Figure 1

26 pages, 1396 KiB  
Review
Dendritic Cell-Based Immunotherapy in Multiple Myeloma: Challenges, Opportunities, and Future Directions
by Emma Verheye, Jesús Bravo Melgar, Sofie Deschoemaeker, Geert Raes, Anke Maes, Elke De Bruyne, Eline Menu, Karin Vanderkerken, Damya Laoui and Kim De Veirman
Int. J. Mol. Sci. 2022, 23(2), 904; https://doi.org/10.3390/ijms23020904 - 14 Jan 2022
Cited by 23 | Viewed by 5322
Abstract
Immunotherapeutic approaches, including adoptive cell therapy, revolutionized treatment in multiple myeloma (MM). As dendritic cells (DCs) are professional antigen-presenting cells and key initiators of tumor-specific immune responses, DC-based immunotherapy represents an attractive therapeutic approach in cancer. The past years, various DC-based approaches, using [...] Read more.
Immunotherapeutic approaches, including adoptive cell therapy, revolutionized treatment in multiple myeloma (MM). As dendritic cells (DCs) are professional antigen-presenting cells and key initiators of tumor-specific immune responses, DC-based immunotherapy represents an attractive therapeutic approach in cancer. The past years, various DC-based approaches, using particularly ex-vivo-generated monocyte-derived DCs, have been tested in preclinical and clinical MM studies. However, long-term and durable responses in MM patients were limited, potentially attributed to the source of monocyte-derived DCs and the immunosuppressive bone marrow microenvironment. In this review, we briefly summarize the DC development in the bone marrow niche and the phenotypical and functional characteristics of the major DC subsets. We address the known DC deficiencies in MM and give an overview of the DC-based vaccination protocols that were tested in MM patients. Lastly, we also provide strategies to improve the efficacy of DC vaccines using new, improved DC-based approaches and combination therapies for MM patients. Full article
(This article belongs to the Special Issue Dendritic Cell and Cancer Therapy)
Show Figures

Figure 1

21 pages, 1471 KiB  
Review
Dendritic Cells and Cancer Immunotherapy: The Adjuvant Effect
by Sara Nava, Daniela Lisini, Simona Frigerio and Anna Bersano
Int. J. Mol. Sci. 2021, 22(22), 12339; https://doi.org/10.3390/ijms222212339 - 15 Nov 2021
Cited by 21 | Viewed by 3611
Abstract
Dendritic cells (DCs) are immune specialized cells playing a critical role in promoting immune response against antigens, and may represent important targets for therapeutic interventions in cancer. DCs can be stimulated ex vivo with pro-inflammatory molecules and loaded with tumor-specific antigen(s). Protocols describing [...] Read more.
Dendritic cells (DCs) are immune specialized cells playing a critical role in promoting immune response against antigens, and may represent important targets for therapeutic interventions in cancer. DCs can be stimulated ex vivo with pro-inflammatory molecules and loaded with tumor-specific antigen(s). Protocols describing the specific details of DCs vaccination manufacturing vary widely, but regardless of the employed protocol, the DCs vaccination safety and its ability to induce antitumor responses is clearly established. Many years of studies have focused on the ability of DCs to provide overall survival benefits at least for a selection of cancer patients. Lessons learned from early trials lead to the hypothesis that, to improve the efficacy of DCs-based immunotherapy, this should be combined with other treatments. Thus, the vaccine’s ultimate role may lie in the combinatorial approaches of DCs-based immunotherapy with chemotherapy and radiotherapy, more than in monotherapy. In this review, we address some key questions regarding the integration of DCs vaccination with multimodality therapy approaches for cancer treatment paradigms. Full article
(This article belongs to the Special Issue Dendritic Cell and Cancer Therapy)
Show Figures

Figure 1

Back to TopTop