1
|
Spiller V, Vetter M, Dettmer-Richardt C, Grammel T. Prospective study of successful autologous dendritic cell therapy in dogs with splenic stage II hemangiosarcoma. Vet J 2024; 306:106196. [PMID: 39004264 DOI: 10.1016/j.tvjl.2024.106196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 07/16/2024]
Abstract
Hemangiosarcoma is an aggressive tumour that most frequently occurs in larger, middle-aged dogs of certain breeds. The spleen is the most commonly affected organ. The aim of this prospective therapy study was to evaluate the clinical effect of autologous, monocyte-derived dendritic cell (DC) therapy in canine hemangiosarcoma stage II after splenectomy. Dogs (n=452) diagnosed with splenic hemangiosarcoma that underwent splenectomy were enrolled. Of these, 42 dogs with stage II entered the DC therapy study. The median survival time for the total group of 42 dogs was 203 days. The median survival for the group (n=34) that received the full DC therapy (≥3 vaccines) was 256 days, with a 29 % one-year survival rate and a hazard ratio of 0.30, adjusted to age and bodyweight (P=0.010). We further observed a significant increase in DC yield after each application and demonstrated that DC yield at the beginning of treatment is significantly related to patient survival. While further evidence is needed, we conclude that autologous, monocyte-derived DC therapy is a viable alternative to standard treatment methods of canine splenic stage II hemangiosarcoma.
Collapse
Affiliation(s)
- V Spiller
- PetBioCell GmbH, Schillerstr. 17, Osterode am Harz 37520, Germany
| | - M Vetter
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Ernst-Grube Str. 40, Halle (Saale) 06120, Germany.
| | | | - T Grammel
- Tiergesundheitszentrum Südharz, Schillerstr. 17, Osterode am Harz 37520, Germany
| |
Collapse
|
2
|
Li P, Jia L, Bian X, Tan S. Application of Engineered Dendritic Cell Vaccines in Cancer Immunotherapy: Challenges and Opportunities. Curr Treat Options Oncol 2023; 24:1703-1719. [PMID: 37962824 DOI: 10.1007/s11864-023-01143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2023] [Indexed: 11/15/2023]
Abstract
OPINION STATEMENT The primary objective of this study is to evaluate the effectiveness of cancer vaccines containing genetically modified dendritic cells (DCs) in inducing transformational immune responses. This paper sheds considerable light on DCs' function in advancing treatment techniques. This objective is achieved by thoroughly analyzing the many facets of DCs and their strategic integration into cancer treatment. Due to their role as immune response regulators, DCs can potentially enhance cancer treatment strategies. DCs have the potential to revolutionize immunotherapy, as shown by a comprehensive analysis of their numerous characteristics. The review deftly transitions from examining the fundamentals of preclinical research to delving into the complexities of clinical implementation while acknowledging the inherent challenges in translating DC vaccine concepts into tangible progress. The analysis also emphasizes the potential synergistic outcomes that can be achieved by combining DC vaccines with established pharmaceuticals, thereby emphasizing the importance of employing a holistic approach to enhance treatment efficacy. Despite the existence of transformative opportunities, advancement is hindered by several obstacles. The exhaustive analysis of technical complexities, regulatory dynamics, and upcoming challenges provides valuable insights for overcoming obstacles requiring strategic navigation to incorporate DC vaccines successfully. This document provides a comprehensive analysis of the developments in DC-based immunotherapy, concentrating on its potential to transform cancer therapy radically.
Collapse
Affiliation(s)
- Ping Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Linan Jia
- Department of Urology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Xiaobo Bian
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang110004, China
| | - Shutao Tan
- Department of Urology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
3
|
Hawlina S, Zorec R, Chowdhury HH. Potential of Personalized Dendritic Cell-Based Immunohybridoma Vaccines to Treat Prostate Cancer. Life (Basel) 2023; 13:1498. [PMID: 37511873 PMCID: PMC10382052 DOI: 10.3390/life13071498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed cancer and the second most common cause of death due to cancer. About 30% of patients with PCa who have been castrated develop a castration-resistant form of the disease (CRPC), which is incurable. In the last decade, new treatments that control the disease have emerged, slowing progression and spread and prolonging survival while maintaining the quality of life. These include immunotherapies; however, we do not yet know the optimal combination and sequence of these therapies with the standard ones. All therapies are not always suitable for every patient due to co-morbidities or adverse effects of therapies or both, so there is an urgent need for further work on new therapeutic options. Advances in cancer immunotherapy with an immune checkpoint inhibition mechanism (e.g., ipilimumab, an anti-CTLA-4 inhibitor) have not shown a survival benefit in patients with CRPC. Other immunological approaches have also not given clear results, which has indirectly prevented breakthrough for this type of therapeutic strategy into clinical use. Currently, the only approved form of immunotherapy for patients with CRPC is a cell-based medicine, but it is only available to patients in some parts of the world. Based on what was gained from recently completed clinical research on immunotherapy with dendritic cell-based immunohybridomas, the aHyC dendritic cell vaccine for patients with CRPC, we highlight the current status and possible alternatives that should be considered in the future.
Collapse
Affiliation(s)
- Simon Hawlina
- Clinical Department of Urology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Surgery, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Helena H Chowdhury
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
4
|
Bloemendal M, Bol KF, Boudewijns S, Gorris MA, de Wilt JH, Croockewit SA, van Rossum MM, de Goede AL, Petry K, Koornstra RH, Figdor C, Gerritsen WR, Schreibelt G, de Vries IJM. Immunological responses to adjuvant vaccination with combined CD1c + myeloid and plasmacytoid dendritic cells in stage III melanoma patients. Oncoimmunology 2021; 11:2015113. [PMID: 36524210 PMCID: PMC9746622 DOI: 10.1080/2162402x.2021.2015113] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We evaluated the immunological responses of lymph-node involved (stage III) melanoma patients to adjuvant dendritic cell vaccination with subsets of naturally occurring dendritic cells (nDCs). Fifteen patients with completely resected stage III melanoma were randomized to receive adjuvant dendritic cell vaccination with CD1c+ myeloid dendritic cells (cDC2s), plasmacytoid dendritic cells (pDCs) or the combination. Immunological response was the primary endpoint and secondary endpoints included safety and survival. In 80% of the patients, antigen-specific CD8+ T cells were detected in skin test-derived T cells and in 55% of patients, antigen-specific CD8+ T cells were detectable in peripheral blood. Functional interferon-γ-producing T cells were found in the skin test of 64% of the patients. Production of nDC vaccines meeting release criteria was feasible for all patients. Vaccination only induced grade 1-2 adverse events, mainly consisting of fatigue. In conclusion, adjuvant dendritic cell vaccination with cDC2s and/or pDCs is feasible, safe and induced immunological responses in the majority of stage III melanoma patients.
Collapse
Affiliation(s)
- Martine Bloemendal
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, the Netherlands,Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kalijn F. Bol
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, the Netherlands,Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Steve Boudewijns
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, the Netherlands,Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mark A.J. Gorris
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, the Netherlands
| | | | | | | | - Anna L. de Goede
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Katja Petry
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Rutger H.T. Koornstra
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Carl Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, the Netherlands,Oncode Institute, Utrecht, the Netherlands
| | - Winald R. Gerritsen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, the Netherlands
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, the Netherlands,CONTACT I. Jolanda M. de Vries Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
5
|
Zhang XW, Huck K, Jähne K, Cichon F, Sonner JK, Ufer F, Bauer S, Woo MS, Green E, Lu K, Kilian M, Friese MA, Platten M, Sahm K. Activity-regulated cytoskeleton-associated protein/activity-regulated gene 3.1 (Arc/Arg3.1) enhances dendritic cell vaccination in experimental melanoma. Oncoimmunology 2021; 10:1920739. [PMID: 34026332 PMCID: PMC8128181 DOI: 10.1080/2162402x.2021.1920739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Dendritic cell (DC) vaccination has proven to be an effective and safe adjuvant for cancer immunotherapies. As the presence of DCs within the tumor microenvironment promotes adaptive antitumor immunity, enhancement of DC migration toward the tumor microenvironment following DC vaccination might represent one possible approach to increase its therapeutic efficacy. While recent findings suggest the activity-regulated cytoskeleton-associated protein/activity-regulated gene 3.1 (Arc/Arg3.1) as critical regulator of DC migration in the context of autoimmune diseases, we aimed to investigate the impact of Arc/Arg3.1 expression for DC-based cancer vaccines. To this end, DC migration capacity as well as the induction of T cell-mediated antitumor immunity was assessed in an experimental B16 melanoma model with Arc/Arg3.1−/- and Arc/Arg3.1-expressing BMDCs applied as a subcutaneous vaccine. While antigen presentation on DCs was critical for unleashing effective T cell mediated antitumor immune responses, Arc/Arg3.1 expression enhanced DC migration toward the tumor and secondary lymphoid organs. Moreover, Arc/Arg3.1-expressing BMDCs shape the tumor immune microenvironment by facilitating tumor recruitment of antigen-specific effector T cells. Thus, Arc/Arg3.1 may represent a novel therapeutic target in DCs in order to increase the therapeutic efficacy of DC vaccination.
Collapse
Affiliation(s)
- Xin-Wen Zhang
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Katrin Huck
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Kristine Jähne
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Frederik Cichon
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Jana K Sonner
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Friederike Ufer
- Institute of Neuroimmunology Und Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Bauer
- Institute of Neuroimmunology Und Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcel Seungsu Woo
- Institute of Neuroimmunology Und Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ed Green
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Kevin Lu
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Michael Kilian
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology Und Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Platten
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Katharina Sahm
- Department of Neurology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
6
|
Tulchiner G, Pichler R, Ulmer H, Staudacher N, Lindner AK, Brunner A, Zelger B, Steinkohl F, Aigner F, Horninger W, Thurnher M. Sex-specific hormone changes during immunotherapy and its influence on survival in metastatic renal cell carcinoma. Cancer Immunol Immunother 2021; 70:2805-2817. [PMID: 33646368 PMCID: PMC8423679 DOI: 10.1007/s00262-021-02882-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022]
Abstract
Renal cell carcinoma (RCC) is a highly vascularized and immunogenic tumor, being an ideal candidate for checkpoint blockade-based immunotherapy. Accordingly, checkpoint inhibitors have demonstrated clinical efficacy in patients with metastatic RCC (mRCC). Sex-specific differences in cancer immunotherapy may be explained by the interaction of sex hormone signaling, genetic and environmental factors, affecting the innate and adaptive immune response in men and women in different ways. The aim of this prospective study was to monitor for the first time changes in sex hormones including luteinizing hormone (LH), follicle-stimulating hormone (FSH), LH/FSH ratio and 17-ß-estradiol (E2) in 22 mRCC patients (12 male and 10 female) receiving nivolumab therapy. In contrast to female patients, male patients showed a significant increase in E2 (p = 0.006) and LH/FSH ratio (p = 0.013) from the beginning of nivolumab therapy to week 12 of follow-up. Moreover, survival analysis revealed a significant negative association between LH/FSH ratio and progression-free survival (PFS) (p = 0.022) as well as between therapy response (p = 0.009) in males compared to females at interim evaluation (week 6/8). Our findings may therefore be the first reference to sex hormone changes during immunotherapy.
Collapse
Affiliation(s)
- Gennadi Tulchiner
- Department of Urology & Immunotherapy Unit, Medical University Innsbruck, Anichstrasse 35 and Innrain 66a, 6020, Innsbruck, Austria
| | - Renate Pichler
- Department of Urology & Immunotherapy Unit, Medical University Innsbruck, Anichstrasse 35 and Innrain 66a, 6020, Innsbruck, Austria.
| | - Hanno Ulmer
- Department of Medical Statistics, Informatics and Health Economics, Medical University Innsbruck, Schoepfstraße 41, 6020, Innsbruck, Austria
| | - Nina Staudacher
- Department of Urology & Immunotherapy Unit, Medical University Innsbruck, Anichstrasse 35 and Innrain 66a, 6020, Innsbruck, Austria
| | - Andrea Katharina Lindner
- Department of Urology & Immunotherapy Unit, Medical University Innsbruck, Anichstrasse 35 and Innrain 66a, 6020, Innsbruck, Austria
| | - Andrea Brunner
- Department of Pathology, Medical University Innsbruck, Muellerstraße 44, 6020, Innsbruck, Austria
| | - Bettina Zelger
- Department of Pathology, Medical University Innsbruck, Muellerstraße 44, 6020, Innsbruck, Austria
| | - Fabian Steinkohl
- Department of Radiology, Medical University Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Friedrich Aigner
- Department of Radiology, Medical University Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Wolfgang Horninger
- Department of Urology & Immunotherapy Unit, Medical University Innsbruck, Anichstrasse 35 and Innrain 66a, 6020, Innsbruck, Austria
| | - Martin Thurnher
- Department of Urology & Immunotherapy Unit, Medical University Innsbruck, Anichstrasse 35 and Innrain 66a, 6020, Innsbruck, Austria.
| |
Collapse
|
7
|
Eisendle K, Weinlich G, Ebner S, Forstner M, Reider D, Zelle‐Rieser C, Tripp CH, Fritsch P, Stoitzner P, Romani N, Nguyen VA. Combining chemotherapy and autologous peptide-pulsed dendritic cells provides survival benefit in stage IV melanoma patients. J Dtsch Dermatol Ges 2020; 18:1270-1277. [PMID: 33197129 PMCID: PMC7756560 DOI: 10.1111/ddg.14334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES We examined retrospectively whether the combination of standard dacarbazine (DTIC) and/or fotemustine chemotherapy and autologous peptide-loaded dendritic cell (DC) vaccination may improve survival of stage IV melanoma patients. Furthermore, a small cohort of long-term survivors was studied in more detail. PATIENTS AND METHODS Between 1998 and 2008, 41 patients were vaccinated at least three times with DCs while receiving chemotherapy and compared to all other 168 patients in our database who only received chemotherapy (1993-2008). RESULTS Median life expectancy of patients receiving additional DC-vaccination was 18 months, compared to eleven months for patients under standard chemotherapy alone. In contrast to patients with other haplotypes, the HLA-A1/A1 subset of DC-treated patients showed significantly lower median survival (12 vs. 25 months). Autoantibodies were frequently detected in serum of both vaccinated and non-vaccinated patients, and there was no correlation between titers, loss or appearance of autoantibodies and survival. Additionally, phenotyping of DCs and PBMCs also did not reveal any conspicuous correlation with survival. CONCLUSIONS Combining standard chemotherapy and DC vaccination appears superior to chemotherapy alone. The impact of HLA haplotypes on survival emphasizes the importance of a careful selection of patients with specific, well-defined HLA haplotypes for future vaccination trials using peptide-pulsed DCs, possibly combined with checkpoint inhibitors.
Collapse
Affiliation(s)
- Klaus Eisendle
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
- Department of Dermatology and VenerologyCentral Hospital of BolzanoItaly
| | - Georg Weinlich
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Susanne Ebner
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
- Department of VisceralTransplant and Thoracic SurgeryMedical University of InnsbruckInnsbruckAustria
| | - Markus Forstner
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Daniela Reider
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Claudia Zelle‐Rieser
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Christoph H. Tripp
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Peter Fritsch
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Patrizia Stoitzner
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Nikolaus Romani
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Van Anh Nguyen
- Department of DermatologyVenereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
8
|
Eisendle K, Weinlich G, Ebner S, Forstner M, Reider D, Zelle‐Rieser C, Tripp CH, Fritsch P, Stoitzner P, Romani N, Nguyen VA. Kombination von Chemotherapie und autologen, Peptid‐beladenen dendritischen Zellen bringt Überlebensvorteil bei Melanompatienten im Stadium IV. J Dtsch Dermatol Ges 2020; 18:1270-1279. [DOI: 10.1111/ddg.14334_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/25/2020] [Indexed: 11/26/2022]
Affiliation(s)
- Klaus Eisendle
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
- Abteilung Dermatologie Venerologie und Allergologie Zentrales Lehrkrankenhaus Bolzano/Bozen Südtiroler Sanitätsbetriebe Bolzano/Bozen Italia
| | - Georg Weinlich
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Susanne Ebner
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
- Universitätsklinik Klinik für Visceral‐ Transplantations‐ und Thoraxchirurgie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Markus Forstner
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Daniela Reider
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Claudia Zelle‐Rieser
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Christoph H. Tripp
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Peter Fritsch
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Patrizia Stoitzner
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Nikolaus Romani
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Van Anh Nguyen
- Universitätsklinik für Dermatologie Venerologie und Allergologie Medizinische Universität Innsbruck Innsbruck Österreich
| |
Collapse
|
9
|
Patel S, Burga RA, Powell AB, Chorvinsky EA, Hoq N, McCormack SE, Van Pelt SN, Hanley PJ, Cruz CRY. Beyond CAR T Cells: Other Cell-Based Immunotherapeutic Strategies Against Cancer. Front Oncol 2019; 9:196. [PMID: 31024832 PMCID: PMC6467966 DOI: 10.3389/fonc.2019.00196] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Chimeric antigen receptor (CAR)-modified T cells have successfully harnessed T cell immunity against malignancies, but they are by no means the only cell therapies in development for cancer. Main Text Summary: Systemic immunity is thought to play a key role in combatting neoplastic disease; in this vein, genetic modifications meant to explore other components of T cell immunity are being evaluated. In addition, other immune cells—from both the innate and adaptive compartments—are in various stages of clinical application. In this review, we focus on these non-CAR T cell immunotherapeutic approaches for malignancy. The first section describes engineering T cells to express non-CAR constructs, and the second section describes other gene-modified cells used to target malignancy. Conclusions: CAR T cell therapies have demonstrated the clinical benefits of harnessing our body's own defenses to combat tumor cells. Similar research is being conducted on lesser known modifications and gene-modified immune cells, which we highlight in this review.
Collapse
Affiliation(s)
- Shabnum Patel
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Rachel A Burga
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Allison B Powell
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Elizabeth A Chorvinsky
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| | - Nia Hoq
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Sarah E McCormack
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Stacey N Van Pelt
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| | - Conrad Russell Y Cruz
- GW Cancer Center, The George Washington University, Washington, DC, United States.,Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| |
Collapse
|
10
|
Martin Lluesma S, Graciotti M, Chiang CLL, Kandalaft LE. Does the Immunocompetent Status of Cancer Patients Have an Impact on Therapeutic DC Vaccination Strategies? Vaccines (Basel) 2018; 6:E79. [PMID: 30477198 PMCID: PMC6313858 DOI: 10.3390/vaccines6040079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/09/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022] Open
Abstract
Although different types of therapeutic vaccines against established cancerous lesions in various indications have been developed since the 1990s, their clinical benefit is still very limited. This observed lack of effectiveness in cancer eradication may be partially due to the often deficient immunocompetent status of cancer patients, which may facilitate tumor development by different mechanisms, including immune evasion. The most frequently used cellular vehicle in clinical trials are dendritic cells (DCs), thanks to their crucial role in initiating and directing immune responses. Viable vaccination options using DCs are available, with a positive toxicity profile. For these reasons, despite their limited therapeutic outcomes, DC vaccination is currently considered an additional immunotherapeutic option that still needs to be further explored. In this review, we propose potential actions aimed at improving DC vaccine efficacy by counteracting the detrimental mechanisms recognized to date and implicated in establishing a poor immunocompetent status in cancer patients.
Collapse
Affiliation(s)
- Silvia Martin Lluesma
- Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Michele Graciotti
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| | - Cheryl Lai-Lai Chiang
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| |
Collapse
|
11
|
Health-related quality of life analysis in stage III melanoma patients treated with adjuvant dendritic cell therapy. Clin Transl Oncol 2018; 21:774-780. [PMID: 30465182 DOI: 10.1007/s12094-018-1987-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/09/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Health-related quality of life (HRQoL) is an important issue in the rapidly evolving field of adjuvant treatment for stage III melanoma. Dendritic cell vaccination is one of the adjuvant forms of therapy currently investigated. METHODS We enrolled adults with stage III melanoma to receive adjuvant dendritic cell vaccination after a complete radical lymph node dissection. HRQoL assessment was one of the secondary endpoints of this trial and investigated with the EORTC-QLQ-C30 questionnaire at baseline and week 26. RESULTS Fifteen patients with a median age of 50 years were included in the study, with twelve evaluable patients on study at time of the second questionnaire. Global health status and role functioning improved clinically relevant with a mean difference of 15 (p = 0.010) and 26 points (p = 0.005), respectively. DISCUSSION Despite the small number of patients, we found a clinically relevant improved global health status. Besides, compared to the other investigated therapies, toxicity of dendritic cell vaccination is low, which supports our finding. CONCLUSION This is the first description of HRQoL in melanoma patients receiving dendritic cell vaccination. We show the expected improvement in global health status after surgical treatment of stage III melanoma. Thus, adjuvant dendritic cell vaccination does not seem to hamper this improvement, as shown in our small explorative study.
Collapse
|
12
|
Hu X, Wu T, Bao Y, Zhang Z. Nanotechnology based therapeutic modality to boost anti-tumor immunity and collapse tumor defense. J Control Release 2017; 256:26-45. [PMID: 28434891 DOI: 10.1016/j.jconrel.2017.04.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 12/19/2022]
Abstract
Cancer is still the leading cause of death. While traditional treatments such as surgery, chemotherapy and radiotherapy play dominating roles, recent breakthroughs in cancer immunotherapy indicate that the influence of immune system on cancer development is virtually beyond our expectation. Manipulating the immune system to fight against cancer has been thriving in recent years. Further understanding of tumor anatomy provides opportunities to put a brake on immunosuppression by overcoming tumor intrinsic resistance or modulating tumor microenvironment. Nanotechnology which provides versatile engineered approaches to enhance therapeutic effects may potentially contribute to the development of future cancer treatment modality. In this review, we will focus on the application of nanotechnology both in boosting anti-tumor immunity and collapsing tumor defense.
Collapse
Affiliation(s)
| | | | - Yuling Bao
- Tongji School of Pharmacy, PR China; Department of Pharmacy, Tongji Hospital, PR China
| | - Zhiping Zhang
- Tongji School of Pharmacy, PR China; National Engineering Research Center for Nanomedicine, PR China; Hubei Engineering Research Center for Novel Drug Delivery System, HuaZhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
13
|
Yoshino S, Nishikawa K, Morita S, Takahashi T, Sakata K, Nagao J, Nemoto H, Murakami N, Matsuda T, Hasegawa H, Shimizu R, Yoshikawa T, Osanai H, Imano M, Naitoh H, Tanaka A, Tajiri T, Gochi A, Suzuki M, Sakamoto J, Saji S, Oka M. Randomised phase III study of S-1 alone versus S-1 plus lentinan for unresectable or recurrent gastric cancer (JFMC36-0701). Eur J Cancer 2016; 65:164-71. [PMID: 27501505 DOI: 10.1016/j.ejca.2016.06.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/17/2016] [Accepted: 06/16/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Lentinan (LNT) is a purified β-1, 3-glucan that augments immune responses. The present study was conducted to assess the efficacy of LNT in combination with S-1 as a first-line treatment for unresectable or recurrent gastric cancer. PATIENTS AND METHODS Eligible patients were randomly assigned to receive S-1 alone or S-1 plus LNT. The primary end-point was overall survival (OS). Secondary end-points were time-to-treatment failure (TTF), overall response rate (ORR), safety, quality of life (QOL), and biomarker. The percentages of LNT-binding monocytes in peripheral blood prior to treatment were analysed for the biomarker assessment. RESULTS One hundred and fifty-four and 155 patients were randomly assigned to receive S-1 alone or S-1 plus LNT, respectively. The median OS was 13.8 and 9.9 months (P = 0.208), the median TTF was 4.3 and 2.6 months (P < 0.001), the ORR was 22.3% and 18.7% for the S-1 and S-1 plus LNT groups, respectively. The incidences of haematologic and non-haematologic adverse events were similar, and no significant changes in QOL scores were observed during the treatment in both groups. In a subpopulation of patients with LNT-binding monocytes ≥2%, patients who received more than two cycles of chemotherapy showed a longer survival time in the S-1 plus LNT group. CONCLUSIONS OS did not improve and TTF was significantly worse in the S-1 plus LNT group as compared with the S-1-only group. This study showed no efficacy of LNT when combined with S-1 treatment in patients with unresectable or recurrent gastric cancer. CLINICAL TRIAL REGISTRATION ID NUMBER UMIN 000000574.
Collapse
Affiliation(s)
- Shigefumi Yoshino
- Department of Digestive Surgery and Surgical Oncology, Yamaguchi University Graduate School of Medicine, Ube, Japan.
| | | | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koichiro Sakata
- Department of Digestive Surgery, Shimonoseki Medical Center, Shimonoseki, Japan
| | - Jiro Nagao
- Department of Surgery, Toho University Omori Medical Center, Tokyo, Japan
| | - Hiroshi Nemoto
- Department of Gastroenterological and General Surgery, School of Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Nozomu Murakami
- Department of Palliative Care and Surgery, Koseiren Takaoka Hospital, Takaoka, Japan
| | - Takeru Matsuda
- Department of Surgery, National Hospital Organization Kobe Medical Center, Kobe, Japan
| | | | - Ryoichi Shimizu
- Department of Surgery, Ogori Daiichi General Hospital, Yamaguchi, Japan
| | - Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Hiroyuki Osanai
- Department of Surgery, Sapporo Geka Kinen Hospital, Sapporo, Japan
| | - Motohiro Imano
- Department of Surgery, Kinki University Faculty of Medicine, Osakasayama, Japan
| | - Hiroshi Naitoh
- Department of Surgery, Japan Community Health Care Organization Gunma Chuo Hospital, Maebashi, Japan
| | | | | | - Akira Gochi
- Digestive Tract Surgery, Okayama University Hospital, Okayama, Japan
| | - Michinari Suzuki
- Department of Surgery, Shunan City Shinnanyo Hospital, Shunan, Japan
| | - Junichi Sakamoto
- Japanese Foundation for Multidisciplinary Treatment of Cancer, Tokyo, Japan
| | - Shigetoyo Saji
- Japanese Foundation for Multidisciplinary Treatment of Cancer, Tokyo, Japan
| | | |
Collapse
|
14
|
Murphy KA, James BR, Guan Y, Torry DS, Wilber A, Griffith TS. Exploiting natural anti-tumor immunity for metastatic renal cell carcinoma. Hum Vaccin Immunother 2016; 11:1612-20. [PMID: 25996049 DOI: 10.1080/21645515.2015.1035849] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Clinical observations of spontaneous disease regression in some renal cell carcinoma (RCC) patients implicate a role for tumor immunity in controlling this disease. Puzzling, however, are findings that high levels of tumor infiltrating lymphocytes (TIL) are common to RCC. Despite expression of activation markers by TILs, functional impairment of innate and adaptive immune cells has been consistently demonstrated contributing to the failure of the immune system to control RCC. Immunotherapy can overcome the immunosuppressive effects of the tumor and provide an opportunity for long-term disease free survival. Unfortunately, complete response rates remain sub-optimal indicating the effectiveness of immunotherapy remains limited by tumor-specific factors and/or cell types that inhibit antitumor immune responses. Here we discuss immunotherapies and the function of multiple immune system components to achieve an effective response. Understanding these complex interactions is essential to rationally develop novel therapies capable of renewing the immune system's ability to respond to these tumors.
Collapse
Affiliation(s)
- Katherine A Murphy
- a Department of Urology; University of Minnesota ; Minneapolis , MN , USA
| | | | | | | | | | | |
Collapse
|
15
|
Martin Lluesma S, Wolfer A, Harari A, Kandalaft LE. Cancer Vaccines in Ovarian Cancer: How Can We Improve? Biomedicines 2016; 4:biomedicines4020010. [PMID: 28536377 PMCID: PMC5344251 DOI: 10.3390/biomedicines4020010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 04/15/2016] [Accepted: 04/19/2016] [Indexed: 12/11/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is one important cause of gynecologic cancer-related death. Currently, the mainstay of ovarian cancer treatment consists of cytoreductive surgery and platinum-based chemotherapy (introduced 30 years ago) but, as the disease is usually diagnosed at an advanced stage, its prognosis remains very poor. Clearly, there is a critical need for new treatment options, and immunotherapy is one attractive alternative. Prophylactic vaccines for prevention of infectious diseases have led to major achievements, yet therapeutic cancer vaccines have shown consistently low efficacy in the past. However, as they are associated with minimal side effects or invasive procedures, efforts directed to improve their efficacy are being deployed, with Dendritic Cell (DC) vaccination strategies standing as one of the more promising options. On the other hand, recent advances in our understanding of immunological mechanisms have led to the development of successful strategies for the treatment of different cancers, such as immune checkpoint blockade strategies. Combining these strategies with DC vaccination approaches and introducing novel combinatorial designs must also be considered and evaluated. In this review, we will analyze past vaccination methods used in ovarian cancer, and we will provide different suggestions aiming to improve their efficacy in future trials.
Collapse
Affiliation(s)
- Silvia Martin Lluesma
- Center of Experimental Therapeutics, Ludwig Center for Cancer Res, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Anita Wolfer
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Alexandre Harari
- Center of Experimental Therapeutics, Ludwig Center for Cancer Res, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Ludwig Center for Cancer Res, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Aris M, Barrio MM. Combining immunotherapy with oncogene-targeted therapy: a new road for melanoma treatment. Front Immunol 2015; 6:46. [PMID: 25709607 PMCID: PMC4321613 DOI: 10.3389/fimmu.2015.00046] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/25/2015] [Indexed: 01/21/2023] Open
Abstract
Cutaneous melanoma arises from the malignant transformation of skin melanocytes; its incidence and mortality have been increasing steadily over the last 50 years, now representing 3% of total tumors. Once melanoma metastasizes, prognosis is somber and therapeutic options are limited. However, the discovery of prevalent BRAF mutations in at least 50% of melanoma tumors led to development of BRAF-inhibitors, and other drugs targeting the MAPK pathway including MEK-inhibitors, are changing this reality. These recently approved treatments for metastatic melanoma have made a significant impact on patient survival; though the results are shadowed by the appearance of drug-resistance. Combination therapies provide a rational strategy to potentiate efficacy and potentially overcome resistance. Undoubtedly, the last decade has also born a renaissance of immunotherapy, and encouraging advances in metastatic melanoma treatment are illuminating the road. Immune checkpoint blockades, such as CTLA-4 antagonist-antibodies, and multiple cancer vaccines are now invaluable arms of anti-tumor therapy. Recent work has brought to light the delicate relationship between tumor biology and the immune system. Host immunity contributes to the anti-tumor activity of oncogene-targeted inhibitors within a complex network of cytokines and chemokines. Therefore, combining immunotherapy with oncogene-targeted drugs may be the key to melanoma control. Here, we review ongoing clinical studies of combination therapies using both oncogene inhibitors and immunotherapeutic strategies in melanoma patients. We will revisit the preclinical evidence that tested sequential and concurrent schemes in suitable animal models and formed the basis for the current trials. Finally, we will discuss potential future directions of the field.
Collapse
Affiliation(s)
- Mariana Aris
- Centro de Investigaciones Oncológicas-Fundación Cáncer , Buenos Aires , Argentina
| | - María Marcela Barrio
- Centro de Investigaciones Oncológicas-Fundación Cáncer , Buenos Aires , Argentina
| |
Collapse
|
17
|
Mody N, Dubey S, Sharma R, Agrawal U, Vyas SP. Dendritic cell-based vaccine research against cancer. Expert Rev Clin Immunol 2014; 11:213-32. [DOI: 10.1586/1744666x.2015.987663] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
18
|
Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN. Clinical use of dendritic cells for cancer therapy. Lancet Oncol 2014; 15:e257-67. [PMID: 24872109 DOI: 10.1016/s1470-2045(13)70585-0] [Citation(s) in RCA: 537] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since the mid-1990s, dendritic cells have been used in clinical trials as cellular mediators for therapeutic vaccination of patients with cancer. Dendritic cell-based immunotherapy is safe and can induce antitumour immunity, even in patients with advanced disease. However, clinical responses have been disappointing, with classic objective tumour response rates rarely exceeding 15%. Paradoxically, findings from emerging research indicate that dendritic cell-based vaccination might improve survival, advocating implementation of alternative endpoints to assess the true clinical potency of dendritic cell-based vaccination. We review the clinical effectiveness of dendritic cell-based vaccine therapy in melanoma, prostate cancer, malignant glioma, and renal cell carcinoma, and summarise the most important lessons from almost two decades of clinical studies of dendritic cell-based immunotherapy in these malignant disorders. We also address how the specialty is evolving, and which new therapeutic concepts are being translated into clinical trials to leverage the clinical effectiveness of dendritic cell-based cancer immunotherapy. Specifically, we discuss two main trends: the implementation of the next-generation dendritic cell vaccines that have improved immunogenicity, and the emerging paradigm of combination of dendritic cell vaccination with other cancer therapies.
Collapse
Affiliation(s)
- Sébastien Anguille
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.
| | - Evelien L Smits
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research, University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Viggo F van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Zwi N Berneman
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| |
Collapse
|
19
|
Bakdash G, Schreurs I, Schreibelt G, Tel J. Crosstalk between dendritic cell subsets and implications for dendritic cell-based anticancer immunotherapy. Expert Rev Clin Immunol 2014; 10:915-26. [PMID: 24758519 DOI: 10.1586/1744666x.2014.912561] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Dendritic cells (DCs) are a family of professional antigen-presenting cells that have an indispensable role in the initiation of innate and adaptive immune responses against pathogens and tumor cells. The DC family is very heterogeneous. Two main types of naturally occurring DCs circulate in peripheral blood, each with its unique phenotypic and functional characteristics: myeloid DCs and plasmacytoid. There is an ample number of studies that have focused on the bi-directional crosstalk between DCs and natural killer cells or T cells. However, the crosstalk among the different DC subsets, in the context of infectious diseases and cancer, has until now not received much attention. Here, we review all available literature that has dealt with the crosstalk between plasmacytoid and myeloid DCs and the potential mode of action. Emphasis will be given to the therapeutic potential of the combination of DC subsets for DC-based immunotherapy.
Collapse
Affiliation(s)
- Ghaith Bakdash
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
20
|
Wang J, Liao L, Tan J. Dendritic cell-based vaccination for renal cell carcinoma: challenges in clinical trials. Immunotherapy 2013; 4:1031-42. [PMID: 23148755 DOI: 10.2217/imt.12.107] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After decades of research, dendritic cell (DC)-based vaccines for renal cell carcinoma have progressed from preclinical rodent models and safety assessments to Phase I/II clinical trials. DC vaccines represent a promising therapy that has produced measurable immunological responses and prolonged survival rates. However, there is still much room to improve in terms of therapeutic efficacy. The key issues that affect the efficiency and reliability of DC therapy include the selection of patients who will respond best to treatment, the proper preparation and administration of DC vaccines, and a combination of DC vaccination with other immune-enhancing therapies (e.g., removal of Tregs, CTLA-4 blockade and lymphodepletion). Additional antiangiogenic agents will hopefully lead to greater survival benefits for patients in early disease stages. This review focuses on the different approaches of DC-based vaccination against renal cell carcinoma and potential strategies to enhance the efficacy of DC vaccination.
Collapse
Affiliation(s)
- Jin Wang
- Organ Transplant Institute, Fuzhou General Hospital, Xiamen University, Fuzhou, China
| | | | | |
Collapse
|
21
|
Tabarkiewicz J. Dendritic cells: active and passive players in therapy of human diseases. Immunotherapy 2012; 4:975-8. [PMID: 23148747 DOI: 10.2217/imt.12.97] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|