1
|
Tiwari A, Alcover K, Carpenter E, Thomas K, Krum J, Nissen A, Van Decar S, Smolinsky T, Valdera F, Vreeland T, Lacher M, Del Priore G, Williams W, Stojadinovic A, Peoples G, Clifton G. Utility of cell-based vaccines as cancer therapy: Systematic review and meta-analysis. Hum Vaccin Immunother 2024; 20:2323256. [PMID: 38544385 PMCID: PMC10984131 DOI: 10.1080/21645515.2024.2323256] [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: 09/16/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Cell-based therapeutic cancer vaccines use autologous patient-derived tumor cells, allogeneic cancer cell lines or autologous antigen presenting cells to mimic the natural immune process and stimulate an adaptive immune response against tumor antigens. The primary objective of this study is to perform a systematic literature review with an embedded meta-analysis of all published Phase 2 and 3 clinical trials of cell-based cancer vaccines in human subjects. The secondary objective of this study is to review trials demonstrating biological activity of cell-based cancer vaccines that could uncover additional hypotheses, which could be used in the design of future studies. We performed the systematic review and meta-analysis according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The final review included 36 studies - 16 single-arm studies, and 20 controlled trials. Our systematic review of the existing literature revealed largely negative trials and our meta-analysis did not show evidence of clinical benefit from cell-based cancer-vaccines. However, as we looked beyond the stringent inclusion criteria of our systematic review, we identified significant examples of biological activity of cell-based cancer vaccines that are worth highlighting. In conclusion, the existing literature on cell-based cancer vaccines is highly variable in terms of cancer type, vaccine therapies and the clinical setting with no overall statistically significant clinical benefit, but there are individual successes that represent the promise of this approach. As cell-based vaccine technology continues to evolve, future studies can perhaps fulfill the potential that this exciting field of anti-cancer therapy holds.
Collapse
Affiliation(s)
- Ankur Tiwari
- Department of Surgery, University of Texas Health Science Center, San Antonio, TX, USA
| | - Karl Alcover
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Katryna Thomas
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | - Julia Krum
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | - Alexander Nissen
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | - Spencer Van Decar
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | - Todd Smolinsky
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | - Franklin Valdera
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | - Timothy Vreeland
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | | | | | | | | | | | - Guy Clifton
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| |
Collapse
|
2
|
Chaoul N, Lauricella E, Giglio A, D'Angelo G, Ganini C, Cives M, Porta C. The future of cellular therapy for the treatment of renal cell carcinoma. Expert Opin Biol Ther 2024; 24:1245-1259. [PMID: 39485013 DOI: 10.1080/14712598.2024.2418321] [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: 06/13/2024] [Revised: 08/26/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024]
Abstract
INTRODUCTION Systemic treatment options for renal cell carcinoma (RCC) have expanded considerably in recent years, and both tyrosine kinase inhibitors and immune checkpoint inhibitors, alone or in combination, have entered the clinical arena. Adoptive cell immunotherapies have recently revolutionized the treatment of cancer and hold the promise to further advance the treatment of RCC. AREAS COVERED In this review, we summarize the latest preclinical and clinical development in the field of adoptive cell immunotherapy for the treatment of RCC, focusing on lymphokine-activated killer (LAK) cells, cytokine-induced killer (CIK) cells, tumor-infiltrating T cells (TILs), TCR-engineered T cells, chimeric antigen receptor (CAR) T cells, and dendritic cell vaccination strategies. Perspectives on emerging cellular products including CAR NK cells, CAR macrophages, as well as γδ T cells are also included. EXPERT OPINION So far, areas of greater therapeutic success of adoptive cell therapies include the adjuvant administration of CIK cells and the transfer of anti-CD70 CAR T cells in patients with metastatic RCC. Bench to bedside and back research will be needed to overcome current limitations of adoptive cell therapies in RCC, primarily aiming at improving the safety of immune cell products, optimizing their antitumor activity and generating off-the-shelf products ready for clinical use.
Collapse
Affiliation(s)
- Nada Chaoul
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Eleonora Lauricella
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Andrea Giglio
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Gabriella D'Angelo
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Carlo Ganini
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Mauro Cives
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Camillo Porta
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| |
Collapse
|
3
|
Dwivedi R, Jain A, Gupta S, Chandra S. Immunotherapy: The Fourth Domain in Oral Cancer Therapeutics. Indian J Otolaryngol Head Neck Surg 2024; 76:2257-2272. [PMID: 38883453 PMCID: PMC11169205 DOI: 10.1007/s12070-024-04565-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/16/2024] [Indexed: 06/18/2024] Open
Abstract
Owing to high global prevalence, incidence and associated mortality, cancer of head and neck particularly oral cancer remains a cardinal domain for research and trials. Immune-modulatory therapies that employ patients own immune system for therapeutic benefits in oral cancer seems promising. The aim of this review is to gauge the potential of immunotherapy as fourth domain of Oral cancer therapeutics. Articles were searched using suitable search terms in MEDLINE and Google Scholar database to include clinical trials, meta-analyses, and research in humans/animals/cell lines published in peer reviewed journals. A total of 97 articles were included in this review. Literature has several studies and trials where different types of immunotherapies has been attempted but it is crucial to identify precise biomarkers of genome based targeted agents and to find parameters to select patients who might benefit from immunotherapy. Also further research is required to estimate predictive value of tumor mutational burden and mutational signatures so as to aid in personalized prediction of oral cancer therapeutic response.
Collapse
Affiliation(s)
- Ruby Dwivedi
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, King George's Medical University, Shahmina Road, Chowk, Lucknow, Uttar Pradesh 226003 India
| | - Ayushi Jain
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, King George's Medical University, Shahmina Road, Chowk, Lucknow, Uttar Pradesh 226003 India
| | - Shalini Gupta
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, King George's Medical University, Shahmina Road, Chowk, Lucknow, Uttar Pradesh 226003 India
| | - Shaleen Chandra
- Atal Bihari Vajpayee Medical University, Lucknow, Uttar Pradesh India
| |
Collapse
|
4
|
Shbeer AM. Current state of knowledge and challenges for harnessing the power of dendritic cells in cancer immunotherapy. Pathol Res Pract 2024; 253:155025. [PMID: 38147726 DOI: 10.1016/j.prp.2023.155025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/28/2023]
Abstract
DCs have great promise for cancer immunotherapy and are essential for coordinating immune responses. In the battle against cancer, using DCs' ability to stimulate the immune system and focus it on tumor cells has shown to be a viable tactic. This study offers a thorough summary of recent developments as well as potential future paths for DC-based immunotherapy against cancer. This study reviews the many methods used in DC therapy, such as vaccination and active cellular immunotherapy. The effectiveness and safety of DC-based treatments for metastatic castration-resistant prostate cancer and non-small cell lung cancer are highlighted in these investigations. The findings indicate longer survival times and superior results for particular patient groups. We are aware of the difficulties and restrictions of DC-based immunotherapy, though. These include the immunosuppressive tumor microenvironment, the intricacy of DC production, and the heterogeneity within DC populations. More study and development are needed to overcome these challenges to enhance immunological responses, optimize treatment regimens, and increase scalability.
Collapse
Affiliation(s)
- Abdullah M Shbeer
- Department of Surgery, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia.
| |
Collapse
|
5
|
Han J, Zhang B, Zheng S, Jiang Y, Zhang X, Mao K. The Progress and Prospects of Immune Cell Therapy for the Treatment of Cancer. Cell Transplant 2024; 33:9636897241231892. [PMID: 38433349 PMCID: PMC10913519 DOI: 10.1177/09636897241231892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 03/05/2024] Open
Abstract
Immune cell therapy as a revolutionary treatment modality, significantly transformed cancer care. It is a specialized form of immunotherapy that utilizes living immune cells as therapeutic reagents for the treatment of cancer. Unlike traditional drugs, cell therapies are considered "living drugs," and these products are currently customized and require advanced manufacturing techniques. Although chimeric antigen receptor (CAR)-T cell therapies have received tremendous attention in the industry regarding the treatment of hematologic malignancies, their effectiveness in treating solid tumors is often restricted, leading to the emergence of alternative immune cell therapies. Tumor-infiltrating lymphocytes (TIL) cell therapy, cytokine-induced killer (CIK) cell therapy, dendritic cell (DC) vaccines, and DC/CIK cell therapy are designed to use the body's natural defense mechanisms to target and eliminate cancer cells, and usually have fewer side effects or risks. On the other hand, cell therapies, such as chimeric antigen receptor-T (CAR-T) cell, T cell receptor (TCR)-T, chimeric antigen receptor-natural killer (CAR-NK), or CAR-macrophages (CAR-M) typically utilize either autologous stem cells, allogeneic or xenogeneic cells, or genetically modified cells, which require higher levels of manipulation and are considered high risk. These high-risk cell therapies typically hold special characteristics in tumor targeting and signal transduction, triggering new anti-tumor immune responses. Recently, significant advances have been achieved in both basic and clinical researches on anti-tumor mechanisms, cell therapy product designs, and technological innovations. With swift technological integration and a high innovation landscape, key future development directions have emerged. To meet the demands of cell therapy technological advancements in treating cancer, we comprehensively and systematically investigate the technological innovation and clinical progress of immune cell therapies in this study. Based on the therapeutic mechanisms and methodological features of immune cell therapies, we analyzed the main technical advantages and clinical transformation risks associated with these therapies. We also analyzed and forecasted the application prospects, providing references for relevant enterprises with the necessary information to make informed decisions regarding their R&D direction selection.
Collapse
Affiliation(s)
- Jia Han
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Bowen Zhang
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Senyu Zheng
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- School of Natural and Computing Sciences, University of Aberdeen, Aberdeen, UK
| | - Yuan Jiang
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Xiaopeng Zhang
- Shanghai World Trade Organization Affairs Consultation Center, Shanghai, China
| | - Kaiyun Mao
- Shanghai Information Center for Life Sciences, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
6
|
Gorodilova AV, Kitaeva KV, Filin IY, Mayasin YP, Kharisova CB, Issa SS, Solovyeva VV, Rizvanov AA. The Potential of Dendritic Cell Subsets in the Development of Personalized Immunotherapy for Cancer Treatment. Curr Issues Mol Biol 2023; 45:8053-8070. [PMID: 37886952 PMCID: PMC10605421 DOI: 10.3390/cimb45100509] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023] Open
Abstract
Since the discovery of dendritic cells (DCs) in 1973 by Ralph Steinman, a tremendous amount of knowledge regarding these innate immunity cells has been accumulating. Their role in regulating both innate and adaptive immune processes is gradually being uncovered. DCs are proficient antigen-presenting cells capable of activating naive T-lymphocytes to initiate and generate effective anti-tumor responses. Although DC-based immunotherapy has not yielded significant results, the substantial number of ongoing clinical trials underscores the relevance of DC vaccines, particularly as adjunctive therapy or in combination with other treatment options. This review presents an overview of current knowledge regarding human DCs, their classification, and the functions of distinct DC populations. The stepwise process of developing therapeutic DC vaccines to treat oncological diseases is discussed, along with speculation on the potential of combined therapy approaches and the role of DC vaccines in modern immunotherapy.
Collapse
Affiliation(s)
- Anna Valerevna Gorodilova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Kristina Viktorovna Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Ivan Yurevich Filin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Yuri Pavlovich Mayasin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Chulpan Bulatovna Kharisova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Shaza S. Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia;
| | - Valeriya Vladimirovna Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Albert Anatolyevich Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| |
Collapse
|
7
|
Oladejo M, Paulishak W, Wood L. Synergistic potential of immune checkpoint inhibitors and therapeutic cancer vaccines. Semin Cancer Biol 2023; 88:81-95. [PMID: 36526110 DOI: 10.1016/j.semcancer.2022.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Cancer vaccines and immune checkpoint inhibitors (ICIs) function at different stages of the cancer immune cycle due to their distinct mechanisms of action. Therapeutic cancer vaccines enhance the activation and infiltration of cytotoxic immune cells into the tumor microenvironment (TME), while ICIs, prevent and/or reverse the dysfunction of these immune cells. The efficacy of both classes of immunotherapy has been evaluated in monotherapy, but they have been met with several challenges. Although therapeutic cancer vaccines can activate anti-tumor immune responses, these responses are susceptible to attenuation by immunoregulatory molecules. Similarly, ICIs are ineffective in the absence of tumor-infiltrating lymphocytes (TILs). Further, ICIs are often associated with immune-related adverse effects that may limit quality of life and compliance. However, the combination of the improved immunogenicity afforded by cancer vaccines and restrained immunosuppression provided by immune checkpoint inhibitors may provide a suitable platform for therapeutic synergism. In this review, we revisit the history and various classifications of therapeutic cancer vaccines. We also provide a summary of the currently approved ICIs. Finally, we provide mechanistic insights into the synergism between ICIs and cancer vaccines.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Wyatt Paulishak
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Laurence Wood
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA.
| |
Collapse
|
8
|
Shi Y, Lu Y, You J. Antigen transfer and its effect on vaccine-induced immune amplification and tolerance. Am J Cancer Res 2022; 12:5888-5913. [PMID: 35966588 PMCID: PMC9373810 DOI: 10.7150/thno.75904] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/15/2022] [Indexed: 12/13/2022] Open
Abstract
Antigen transfer refers to the process of intercellular information exchange, where antigenic components including nucleic acids, antigen proteins/peptides and peptide-major histocompatibility complexes (p-MHCs) are transmitted from donor cells to recipient cells at the thymus, secondary lymphoid organs (SLOs), intestine, allergic sites, allografts, pathological lesions and vaccine injection sites via trogocytosis, gap junctions, tunnel nanotubes (TNTs), or extracellular vesicles (EVs). In the context of vaccine inoculation, antigen transfer is manipulated by the vaccine type and administration route, which consequently influences, even alters the immunological outcome, i.e., immune amplification and tolerance. Mainly focused on dendritic cells (DCs)-based antigen receptors, this review systematically introduces the biological process, molecular basis and clinical manifestation of antigen transfer.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
9
|
Guo J, Zhang Y, Qin Q, Chao N, Huang T, Chen C, Lu X, Huang R, Pan J. Dendritic cells modified by tumor associated antigen SMP30 have enhanced antitumor effect against mouse hepatocarcinoma cells in vitro and in vivo. Am J Transl Res 2022; 14:5785-5799. [PMID: 36105050 PMCID: PMC9452339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES Tumor immunotherapy based on dendritic cells (DC) is one of the most promising approaches to treat cancers. This therapy uses an immunogenic tumor antigen to present it to T cells. Senescence marker protein 30 (SMP30) is identified as a tumor associated antigen (TAA) with high immunogenicity and specificity for hepatocellular carcinoma (HCC). DCs are the most potent antigen presenting cells, and can be transduced with tumor antigens to enhance antitumor immune response. The purpose of this study was to investigate the antitumor effect of DCs transduced with a recombinant lentiviral vector (LV-SMP30) expressing SMP30. METHODS A recombinant lentiviral vector (LV-SMP30) expressing SMP30 was constructed and transduced into DCs. The expression of SMP30 was detected by western blot. Mouse bone marrow-derived DCs were divided into four groups: LV-SMP30 group (transduced with LV-SMP30), Protein group (co-cultured with SMP30 protein), LV group (transduced with the empty vector) and Untreated group (the normal DCs). The effect of LV-SMP30 on DCs was detected through surface markers (CD123, CD11c, CD80 and CD86) and cytokine production. The activation and proliferation of CD3+CD8+ T cells were detected by CCK-8 kit. Flow cytometry was used to detect CD3+CD8+ T cell-mediated cytotoxicity. After construction of a mouse subcutaneous xenograft model, the volume and growth of tumors in different groups were observed. The changes in serum immune indexes in the treated groups were compared with those in the control group. RESULTS The LV-SMP30 recombinant was constructed and transduced into DCs successfully, and LV-SMP30-transduced DCs stably expressed SMP30. The percentages of expression in the LV-SMP30 and Protein groups were significantly higher than those in the LV or Untreated groups (P<0.05). Meanwhile, after the DCs were cultured for 72 hours, the levels of IL-2, IL-6, IL-12, and IFN-γ were significantly higher in the LV-SMP30 and Protein groups than in the LV group or Untreated group (P<0.05). After the DCs were continuously cultured for one week, however, the cytokine levels in the LV-SMP30 group were significantly higher than those in the Protein group (P<0.05). In addition, CD3+CD8+ T cell proliferation and activation levels were substantially higher in the LV-SMP30 and Protein groups than in the LV or Untreated groups (P<0.05). Furthermore, as the ratio of effectors/target cells increasing in the LV-SMP30 group, CD3+CD8+ T cell-mediated cytotoxicity in H22 cells became higher (0:1, 10:1; 20:1; 40:1, respectively). In comparison to the control group, the cytotoxicity of the LV-SMP30 group was considerably increased at the ratios of 10:1, 20:1 and 40:1 (P<0.05). However, in the case of Hep1-6 cells, there was no significant difference in CD3+CD8+ T cell-mediated cytotoxicity among the groups. In addition, when compared with other groups, the mice in the LV-SMP30 group showed the most volume reduction, the slowest tumor growth, and the highest level of IL-2 and IFN-γ (P<0.05). CONCLUSION DCs transduced with LV-SMP30 can dramatically enhance specific CD3+CD8+ T cell immune responses against mouse hepatocarcinoma cells in vitro and in vivo. These findings lend significant support to the development of the DC-based SMP30 antigen vaccine for hepatocarcinoma immunotherapy.
Collapse
Affiliation(s)
- Jinhong Guo
- College of Stomatology, Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center, Guangxi Medical UniversityNanning 530021, Guangxi, P. R. China
| | - Yaoyao Zhang
- Department of Histology and Embryology, Guangxi Medical UniversityNanning 530021, Guangxi, P. R. China
| | - Qiuhong Qin
- Department of Pathology, Jiangbin Hospital of Guangxi Zhuang Autonomous RegionNanning 530021, Guangxi, P. R. China
| | - Naixia Chao
- Department of Histology and Embryology, Guangxi Medical UniversityNanning 530021, Guangxi, P. R. China
| | - Tianming Huang
- Department of Histology and Embryology, Guangxi Medical UniversityNanning 530021, Guangxi, P. R. China
| | - Chengxiao Chen
- Department of Histology and Embryology, Guangxi Medical UniversityNanning 530021, Guangxi, P. R. China
| | - Xiaoling Lu
- College of Stomatology, Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center, Guangxi Medical UniversityNanning 530021, Guangxi, P. R. China
| | - Rongshi Huang
- Department of Histology and Embryology, Institute of Preclinical, Guangxi Traditional Chinese Medical UniversityNanning 530001, Guangxi, P. R. China
| | - Jian Pan
- Department of Human Anatomy, Guangxi Medical UniversityNanning 530021, Guangxi, P. R. China
| |
Collapse
|
10
|
A therapeutic DC vaccine with maintained immunological activity exhibits robust anti-tumor efficacy. J Control Release 2022; 349:254-268. [PMID: 35803328 DOI: 10.1016/j.jconrel.2022.06.059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 11/22/2022]
Abstract
Dendritic cells (DCs) vaccines are a major focus of future anti-tumor immunotherapy for their pivotal role in eliciting reactive tumor-specific T-cell responses. Tumor cell-mediated DCs (TC-DC) activation and tumor antigen-mediated DCs (TA-DC) activation are two conventional modes of DC vaccine construction in clinical studies. The former physiologically mimicks the tumor identification and rejection, significantly contributing to DC-based immune recognition and migration towards the complexed tumor microenvironment (TME). However, as immunosuppressive molecules may exist in TME, these TC-DC are generally characterized with aberrant lipid accumulation and inositol-requiring kinase 1α (IRE1α)-X-box binding protein 1 (XBP1) hyperactivation, which is provoked by overwhelming oxidative stress and endoplasmic reticulum (ER) stress, resulting in TC-DC malfunction. Oppositely, without contacting immunosuppressive TME, TA-DC vaccines perform better in T-cell priming and lymph nodes (LNs) homing, but are relatively weak in TME infiltration and identification. Herein, we prepared a KIRA6-loaded α-Tocopherol nanoemulsion (KT-NE), which simultaneously ameliorated oxidative stress and ER stress in the dysfunctional lipid-laden TC-DC. The TC-DC treated by KT-NE could maintain immunological activity, simultaneously, exhibited satisfactory chemotaxis towards LNs and tumor sites in vivo, and effectively suppressed malignant progression by unleashing activated tumor-reactive T cells. This study generated a new DC-vaccine that owned puissant aptitude to identify complicated TME as well as robust immunological activity to boost T-cell initiation, which may provide some insights into the design and application of DC-vaccines for clinical application.
Collapse
|
11
|
Zhao L, Zhang S, Kepp O, Kroemer G, Liu P. Dendritic cell transfer for cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 370:33-64. [PMID: 35798506 DOI: 10.1016/bs.ircmb.2022.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Dendritic cells (DCs) play a major role in cancer immunosurveillance as they bridge innate and adaptive immunity by detecting tumor-associated antigens and presenting them to T lymphocytes. The adoptive transfer of antigen loaded DCs has been proposed as an immunotherapeutic approach for the treatment of various types of cancer. Nevertheless, despite promising preclinical data, the therapeutic efficacy of DC transfer is still deceptive in cancer patients. Here we summarize recent findings in DC biology with a special focus on the development of actionable therapeutic strategies and discuss experimental and clinical approaches that aim at improving the efficacy of DC-based immunotherapies, including, but not limited to, optimized DC production and antigen loading, stimulated maturation, the co-treatment with additional immunotherapies, as well as the inhibition of DC checkpoints.
Collapse
Affiliation(s)
- Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Shuai Zhang
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Institut du Cancer Paris Carpem, Department of Biology, Hôpital Européen Georges Pompidou, APHP, Paris, France.
| | - Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
| |
Collapse
|
12
|
Thongchot S, Jirapongwattana N, Luangwattananun P, Chiraphapphaiboon W, Chuangchot N, Sa-nguanraksa D, O-Charoenrat P, Thuwajit P, Yenchitsomanus PT, Thuwajit C. Adoptive Transfer of Anti-Nucleolin T Cells Combined with PD-L1 Inhibition against Triple-Negative Breast Cancer. Mol Cancer Ther 2022; 21:727-739. [PMID: 35313339 PMCID: PMC9377762 DOI: 10.1158/1535-7163.mct-21-0823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/21/2021] [Accepted: 03/02/2022] [Indexed: 01/18/2023]
Abstract
Dendritic cell (DC)-based T-cell activation is an alternative immunotherapy in breast cancer. The anti-programmed death ligand 1 (PD-L1) can enhance T-cell function. Nucleolin (NCL) is overexpressed in triple-negative breast cancer (TNBC). The regulation of PD-L1 expression through autophagy and the anti-PD-L1 peptide to help sensitize T cells for NCL-positive TNBC cell killing has not been evaluated. Results showed the worst clinical outcome in patients with high NCL and PD-L1. Self-differentiated myeloid-derived antigen-presenting cells reactive against tumors presenting NCL or SmartDCs-NCL producing GM-CSF and IL-4, could activate NCL-specific T cells. SmartDCs-NCL plus recombinant human ribosomal protein substrate 3 (RPS3) successfully induced maturation and activation of DCs characterized by the reduction of CD14 and the induction of CD11c, CD40, CD80, CD83, CD86, and HLA-DR. Interestingly, SmartDCs-NCL plus RPS3 in combination with anti-PD-L1 peptide revealed significant killing activity of the effector NCL-specific T cells against NCLHigh/PD-L1High MDA-MB-231 and NCLHigh/PD-L1High HCC70 TNBC cells at the effector: a target ratio of 5:1 in 2-D and 10:1 in the 3-D culture system; and increments of IFNγ by the ELISpot assay. No killing effect was revealed in MCF-10A normal mammary cells. Mechanistically, NCL-specific T-cell-mediated TNBC cell killing was through both apoptotic and autophagic pathways. Induction of autophagy by curcumin, an autophagic stimulator, inhibited the expression of PD-L1 and enhanced cytolytic activity of NCL-specific T cells. These findings provide the potential clinical approaches targeting NCLHigh/PD-L1High TNBC cells with NCL-specific T cells in combination with a PD-L1 inhibitor or autophagic stimulator.
Collapse
Affiliation(s)
- Suyanee Thongchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Research Department, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Niphat Jirapongwattana
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Piriya Luangwattananun
- Research Department, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wannasiri Chiraphapphaiboon
- Research Department, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Biochemistry, International Graduate Program in Medical Biochemistry and Molecular Biology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nisa Chuangchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Research Department, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Doonyapat Sa-nguanraksa
- Department of Clinical Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pa-thai Yenchitsomanus
- Research Department, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW The clinical activity of new immunotherapies in cancer, such as anti-Programmed cell death 1 (PD-1)/Programmed death-ligand 1, has revealed the importance of the patient's immune system in controlling tumor development. As in infectious diseases, dendritic cells (DCs) are critical for inducing immune responses in cancer. Unfortunately, autologous DC-based vaccines have not yet demonstrated their clinical benefit. Here, we review recent research using allogeneic DCs as alternatives to autologous DCs to develop innovative therapeutic cancer vaccines. RECENT FINDINGS A novel approach using an allogeneic plasmacytoid dendritic cell (PDC) line as an antigen presentation platform showed great potency when used to prime and expand antitumor-specific CD8+ T cells in vitro and in vivo in a humanized mouse model. This PDC platform, named PDC∗vac, was first evaluated in the treatment of melanoma with encouraging results and is currently being evaluated in the treatment of lung cancer in combination with anti-PD-1 immunotherapy. SUMMARY Therapeutic cancer vaccines are of particular interest because they aim to help patients, to mount effective antitumor responses, especially those who insufficiently respond to immune checkpoint inhibitors. The use of an allogeneic plasmacytoid DC-based platform such as PDC∗vac could greatly potentiate the efficacy of these new immunotherapies.
Collapse
Affiliation(s)
- Joël Plumas
- Immunobiology and Immunotherapy of Chronic Diseases, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, Université Grenoble Alpes
- Research and Development Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes
- PDC∗line Pharma SAS, Grenoble, France
- PDC∗line Pharma SA, Liège, Belgium
| |
Collapse
|
14
|
Lu Y, Shi Y, You J. Strategy and clinical application of up-regulating cross presentation by DCs in anti-tumor therapy. J Control Release 2021; 341:184-205. [PMID: 34774890 DOI: 10.1016/j.jconrel.2021.11.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/20/2022]
Abstract
The cross presentation of exogenous antigen (Ag) by dendritic cells (DCs) facilitates a diversified mode of T-cell activation, orchestrates specific humoral and cellular immunity, and contributes to an efficient anti-tumor immune response. DCs-mediated cross presentation is subject to both intrinsic and extrinsic factors, including the homing and phenotype of DCs, the spatiotemporal trafficking and degradation kinetics of Ag, and multiple microenvironmental clues, with many details largely unexplored. Here, we systemically review the current mechanistic understanding and regulation strategies of cross presentation by heterogeneous DC populations. We also provide insights into the future exploitation of DCs cross presentation for a better clinical efficacy in anti-tumor therapy.
Collapse
Affiliation(s)
- Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
15
|
Chung JY, Thone MN, Kwon YJ. COVID-19 vaccines: The status and perspectives in delivery points of view. Adv Drug Deliv Rev 2021; 170:1-25. [PMID: 33359141 PMCID: PMC7759095 DOI: 10.1016/j.addr.2020.12.011] [Citation(s) in RCA: 217] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/29/2022]
Abstract
Due to the high prevalence and long incubation periods often without symptoms, the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has infected millions of individuals globally, causing the coronavirus disease 2019 (COVID-19) pandemic. Even with the recent approval of the anti-viral drug, remdesivir, and Emergency Use Authorization of monoclonal antibodies against S protein, bamlanivimab and casirimab/imdevimab, efficient and safe COVID-19 vaccines are still desperately demanded not only to prevent its spread but also to restore social and economic activities via generating mass immunization. Recent Emergency Use Authorization of Pfizer and BioNTech's mRNA vaccine may provide a pathway forward, but monitoring of long-term immunity is still required, and diverse candidates are still under development. As the knowledge of SARS-CoV-2 pathogenesis and interactions with the immune system continues to evolve, a variety of drug candidates are under investigation and in clinical trials. Potential vaccines and therapeutics against COVID-19 include repurposed drugs, monoclonal antibodies, antiviral and antigenic proteins, peptides, and genetically engineered viruses. This paper reviews the virology and immunology of SARS-CoV-2, alternative therapies for COVID-19 to vaccination, principles and design considerations in COVID-19 vaccine development, and the promises and roles of vaccine carriers in addressing the unique immunopathological challenges presented by the disease.
Collapse
Affiliation(s)
- Jee Young Chung
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States of America
| | - Melissa N Thone
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States of America
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States of America; Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, United States of America; Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, United States of America; Department of Biomedical Engineering, University of California, Irvine, CA 92697, United States of America.
| |
Collapse
|
16
|
Naseri M, Zöller M, Hadjati J, Ghods R, Ranaei Pirmardan E, Kiani J, Eini L, Bozorgmehr M, Madjd Z. Dendritic cells loaded with exosomes derived from cancer stem cell-enriched spheroids as a potential immunotherapeutic option. J Cell Mol Med 2021; 25:3312-3326. [PMID: 33634564 PMCID: PMC8034455 DOI: 10.1111/jcmm.16401] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/30/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are responsible for therapeutic resistance and recurrence in colorectal cancer. Despite advances in immunotherapy, the inability to specifically eradicate CSCs has led to treatment failure. Hence, identification of appropriate antigen sources is a major challenge in designing dendritic cell (DC)‐based therapeutic strategies against CSCs. Here, in an in vitro model using the HT‐29 colon cancer cell line, we explored the efficacy of DCs loaded with exosomes derived from CSC‐enriched colonospheres (CSCenr‐EXOs) as an antigen source in activating CSC‐specific T‐cell responses. HT‐29 lysate, HT‐29‐EXOs and CSCenr lysate were independently assessed as separate antigen sources. Having confirmed CSCs enrichment in spheroids, CSCenr‐EXOs were purified and characterized, and their impact on DC maturation was investigated. Finally, the impact of the antigen‐pulsed DCs on the proliferation rate and also spheroid destructive capacity of autologous T cells was assessed. CSCenr‐EXOs similar to other antigen groups had no suppressive/negative impacts on phenotypic maturation of DCs as judged by the expression level of costimulatory molecules. Notably, similar to CSCenr lysate, CSCenr‐EXOs significantly increased the IL‐12/IL‐10 ratio in supernatants of mature DCs. CSCenr‐EXO‐loaded DCs effectively promoted T‐cell proliferation. Importantly, T cells stimulated with CSCenr‐EXOs disrupted spheroids' structure. Thus, CSCenr‐EXOs present a novel and promising antigen source that in combination with conventional tumour bulk‐derived antigens should be further explored in pre‐clinical immunotherapeutic settings for the efficacy in hampering recurrence and metastatic spread.
Collapse
Affiliation(s)
- Marzieh Naseri
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Margot Zöller
- Section Pancreas Research, University Hospital of Surgery, Heidelberg, Germany
| | - Jamshid Hadjati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Ehsan Ranaei Pirmardan
- Department of Radiology, Molecular Biomarkers Nano-imaging Laboratory, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Leila Eini
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Basic Science, Faculty of Veterinary, Science and Research Branch of Islamic, Azad University, Tehran, Iran
| | - Mahmood Bozorgmehr
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
17
|
Sheehan C, D'Souza-Schorey C. Tumor-derived extracellular vesicles: molecular parcels that enable regulation of the immune response in cancer. J Cell Sci 2019; 132:132/20/jcs235085. [PMID: 31615844 DOI: 10.1242/jcs.235085] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous collection of membrane-bound vesicles released by cells that contain bioactive cargoes including proteins, lipids and nucleic acids. Multiple subpopulations of EVs have now been recognized and these include exosomes and microvesicles. EVs have been thought to facilitate intercellular and distal communication to bring about various processes that enable tumor progression and metastases. Here, we describe the current knowledge of the functional cargo contained within EVs, with a focus on tumor microvesicles, and review the emerging theory of how EVs support immune suppression in cancer.
Collapse
Affiliation(s)
- Colin Sheehan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556-0369, USA
| | | |
Collapse
|
18
|
Abstract
Immunotherapy using dendritic cell (DC)-based vaccination is an approved approach for harnessing the potential of a patient's own immune system to eliminate tumor cells in metastatic hormone-refractory cancer. Overall, although many DC vaccines have been tested in the clinic and proven to be immunogenic, and in some cases associated with clinical outcome, there remains no consensus on how to manufacture DC vaccines. In this review we will discuss what has been learned thus far about human DC biology from clinical studies, and how current approaches to apply DC vaccines in the clinic could be improved to enhance anti-tumor immunity.
Collapse
|
19
|
Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, Hernandez G, Mier J, He X, Hodi FS, Denker M, Leveque V, Cañamero M, Babitski G, Koeppen H, Ziai J, Sharma N, Gaire F, Chen DS, Waterkamp D, Hegde PS, McDermott DF. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun 2016; 7:12624. [PMID: 27571927 PMCID: PMC5013615 DOI: 10.1038/ncomms12624] [Citation(s) in RCA: 530] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/18/2016] [Indexed: 12/16/2022] Open
Abstract
Anti-tumour immune activation by checkpoint inhibitors leads to durable responses in a variety of cancers, but combination approaches are required to extend this benefit beyond a subset of patients. In preclinical models tumour-derived VEGF limits immune cell activity while anti-VEGF augments intra-tumoral T-cell infiltration, potentially through vascular normalization and endothelial cell activation. This study investigates how VEGF blockade with bevacizumab could potentiate PD-L1 checkpoint inhibition with atezolizumab in mRCC. Tissue collections are before treatment, after bevacizumab and after the addition of atezolizumab. We discover that intra-tumoral CD8+ T cells increase following combination treatment. A related increase is found in intra-tumoral MHC-I, Th1 and T-effector markers, and chemokines, most notably CX3CL1 (fractalkine). We also discover that the fractalkine receptor increases on peripheral CD8+ T cells with treatment. Furthermore, trafficking lymphocyte increases are observed in tumors following bevacizumab and combination treatment. These data suggest that the anti-VEGF and anti-PD-L1 combination improves antigen-specific T-cell migration. Cancer immunotherapy can be used in combination with other therapies for a better response. Here, the authors conduct a phase Ib clinical study and report the clinical activity and the immune response of the anti-PDL1 agent, atezolizumab, in combination with bevacizumab in ten patients with metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Jeffrey J Wallin
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Johanna C Bendell
- GI Oncology Research, Drug Development Unit, Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, Tennessee, 37203, USA
| | - Roel Funke
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Mario Sznol
- Department of Internal Medicine and Melanoma Unit, Yale Cancer Center, New Haven, Connecticut 06511, USA
| | | | - Suzanne Jones
- GI Oncology Research, Drug Development Unit, Sarah Cannon Research Institute, 250 25th Avenue North, Suite 100, Nashville, Tennessee, 37203, USA
| | | | - James Mier
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, Massachusets 02215, USA
| | - Xian He
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - F Stephen Hodi
- Dana-Farber/Brigham and Women's Cancer Center, 450 Brookline Avenue, Boston, Massachusets 02215, USA
| | - Mitchell Denker
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Vincent Leveque
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Marta Cañamero
- Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Galina Babitski
- Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Hartmut Koeppen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - James Ziai
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Neeraj Sharma
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Fabien Gaire
- Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Daniel S Chen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Daniel Waterkamp
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Priti S Hegde
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - David F McDermott
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, Massachusets 02215, USA
| |
Collapse
|
20
|
Lim S, Koo JH, Choi JM. Use of Cell-Penetrating Peptides in Dendritic Cell-Based Vaccination. Immune Netw 2016; 16:33-43. [PMID: 26937230 PMCID: PMC4770098 DOI: 10.4110/in.2016.16.1.33] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 12/13/2022] Open
Abstract
Cell-penetrating peptides (CPPs) are short amino acids that have been widely used to deliver macromolecules such as proteins, peptides, DNA, or RNA, to control cellular behavior for therapeutic purposes. CPPs have been used to treat immunological diseases through the delivery of immune modulatory molecules in vivo. Their intracellular delivery efficiency is highly synergistic with the cellular characteristics of the dendritic cells (DCs), which actively uptake foreign antigens. DC-based vaccines are primarily generated by pulsing DCs ex vivo with various immunomodulatory antigens. CPP conjugation to antigens would increase DC uptake as well as antigen processing and presentation on both MHC class II and MHC class I molecules, leading to antigen specific CD4(+) and CD8(+) T cell responses. CPP-antigen based DC vaccination is considered a promising tool for cancer immunotherapy due to the enhanced CTL response. In this review, we discuss the various applications of CPPs in immune modulation and DC vaccination, and highlight the advantages and limitations of the current CPP-based DC vaccination.
Collapse
Affiliation(s)
- Sangho Lim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea.; Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Ja-Hyun Koo
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea.; Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea.; Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
21
|
Constantino J, Gomes C, Falcão A, Cruz MT, Neves BM. Antitumor dendritic cell-based vaccines: lessons from 20 years of clinical trials and future perspectives. Transl Res 2016; 168:74-95. [PMID: 26297944 DOI: 10.1016/j.trsl.2015.07.008] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/25/2015] [Accepted: 07/28/2015] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are versatile elements of the immune system and are best known for their unparalleled ability to initiate and modulate adaptive immune responses. During the past few decades, DCs have been the subject of numerous studies seeking new immunotherapeutic strategies against cancer. Despite the initial enthusiasm, disappointing results from early studies raised some doubts regarding the true clinical value of these approaches. However, our expanding knowledge of DC immunobiology and the definition of the optimal characteristics for antitumor immune responses have allowed a more rational development of DC-based immunotherapies in recent years. Here, after a brief overview of DC immunobiology, we sought to systematize the knowledge provided by 20 years of clinical trials, with a special emphasis on the diversity of approaches used to manipulate DCs and their consequent impact on vaccine effectiveness. We also address how new therapeutic concepts, namely the combination of DC vaccines with other anticancer therapies, are being implemented and are leveraging clinical outcomes. Finally, optimization strategies, new insights, and future perspectives on the field are also highlighted.
Collapse
Affiliation(s)
- João Constantino
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Célia Gomes
- Faculty of Medicine, Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI) and Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Maria T Cruz
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Bruno M Neves
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal; Department of Chemistry and QOPNA, Mass Spectrometry Centre, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
22
|
Markov OV, Mironova NL, Sennikov SV, Vlassov VV, Zenkova MA. Prophylactic Dendritic Cell-Based Vaccines Efficiently Inhibit Metastases in Murine Metastatic Melanoma. PLoS One 2015; 10:e0136911. [PMID: 26325576 PMCID: PMC4556596 DOI: 10.1371/journal.pone.0136911] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/10/2015] [Indexed: 12/16/2022] Open
Abstract
Recent data on the application of dendritic cells (DCs) as anti-tumor vaccines has shown their great potential in therapy and prophylaxis of cancer. Here we report on a comparison of two treatment schemes with DCs that display the models of prophylactic and therapeutic vaccination using three different experimental tumor models: namely, Krebs-2 adenocarcinoma (primary tumor), melanoma (B16, metastatic tumor without a primary node) and Lewis lung carcinoma (LLC, metastatic tumor with a primary node). Dendritic cells generated from bone marrow-derived DC precursors and loaded with lysate of tumor cells or transfected with the complexes of total tumor RNA with cationic liposomes were used for vaccination. Lipofectamine 2000 and liposomes consisting of helper lipid DOPE (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine) and cationic lipid 2D3 (1,26-Bis(1,2-de-O-tetradecyl-rac-glycerol)-7,11,16,20-tetraazahexacosan tetrahydrocloride) were used for RNA transfection. It was shown that DCs loaded with tumor lysate were ineffective in contrast to tumor-derived RNA. Therapeutic vaccination with DCs loaded by lipoplexes RNA/Lipofectamine 2000 was the most efficient for treatment of non-metastatic Krebs-2, where a 1.9-fold tumor growth retardation was observed. Single prophylactic vaccination with DCs loaded by lipoplexes RNA/2D3 was the most efficient to treat highly aggressive metastatic tumors LLC and B16, where 4.7- and 10-fold suppression of the number of lung metastases was observed, respectively. Antimetastatic effect of single prophylactic DC vaccination in metastatic melanoma model was accompanied by the reductions in the levels of Th2-specific cytokines however the change of the levels of Th1/Th2/Th17 master regulators was not found. Failure of double prophylactic vaccination is explained by Th17-response polarization associated with autoimmune and pro-inflammatory reactions. In the case of therapeutic DC vaccine the polarization of Th1-response was found nevertheless the antimetastatic effect was less effective in comparison with prophylactic DC vaccine.
Collapse
Affiliation(s)
- Oleg V. Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Nadezhda L. Mironova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey V. Sennikov
- Federal State Budgetary Institution "Research Institute of Clinical Immunology", Siberian Branch of Russian Academy of Medical Sciences, Novosibirsk, Russia
| | - Valentin V. Vlassov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
- * E-mail:
| |
Collapse
|
23
|
Harnessing the PD-1 pathway in renal cell carcinoma: current evidence and future directions. BioDrugs 2015; 28:513-26. [PMID: 25445176 DOI: 10.1007/s40259-014-0111-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Programmed cell death-1 (PD-1) is a recognized immune checkpoint. It is frequently upregulated on the T cells that infiltrate tumors, providing an inhibitory signal, which may facilitate immune escape. Blocking antibodies have been developed to interrupt the interaction of PD-1 with its ligands PD-L1/PD-L2, with the goal of increasing the host antitumor immune response. Initial results have been encouraging, with durable responses in both treatment-naive and pretreated patients, along with an acceptable toxicity profile. This tolerability makes PD-1 blockade an excellent potential partner for combination strategies with the approved targeted agents, such as tyrosine kinase inhibitors (TKIs) and anti-vascular endothelial growth factor (anti-VEGF) antibodies, as well as other investigational immune checkpoint inhibitors or agonist antibodies that may costimulate an immune response. PD-L1 expression on tumor cells and tumor-infiltrating immune cells is also being evaluated as a predictive biomarker of response to treatment. This review summarizes the biological basis, preclinical studies, ongoing trials, and future challenges associated with targeting the PD-1 pathway in renal cell carcinoma.
Collapse
|
24
|
Cany J, Dolstra H, Shah N. Umbilical cord blood-derived cellular products for cancer immunotherapy. Cytotherapy 2015; 17:739-748. [PMID: 25795272 DOI: 10.1016/j.jcyt.2015.03.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/03/2015] [Indexed: 11/16/2022]
Abstract
Although the vast majority of experience with umbilical cord blood (CB) centers on hematopoietic reconstitution, a recent surge in the knowledge of CB cell subpopulations as well as advances in ex vivo culture technology have expanded the potential of this rich resource. Because CB has the capacity to generate the entire hematopoietic system, we now have a new source for natural killer, dendritic and T cells for therapeutic use against malignancies. This Review will focus on cellular immunotherapies derived from CB. Expansion techniques, ongoing clinical trials and future directions for this new dimension of CB application are also discussed.
Collapse
Affiliation(s)
- Jeannette Cany
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nina Shah
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
25
|
Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2013; 2:e25771. [PMID: 24286020 PMCID: PMC3841205 DOI: 10.4161/onci.25771] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 12/26/2022] Open
Abstract
Dendritic cells (DCs) occupy a privileged position at the interface between innate and adaptive immunity, orchestrating a large panel of responses to both physiological and pathological cues. In particular, whereas the presentation of antigens by immature DCs generally results in the development of immunological tolerance, mature DCs are capable of priming robust, and hence therapeutically relevant, adaptive immune responses. In line with this notion, functional defects in the DC compartment have been shown to etiologically contribute to pathological conditions including (but perhaps not limited to) infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. Thus, the possibility of harnessing the elevated immunological potential of DCs for anticancer therapy has attracted considerable interest from both researchers and clinicians over the last decade. Alongside, several methods have been developed not only to isolate DCs from cancer patients, expand them, load them with tumor-associated antigens and hence generate highly immunogenic clinical grade infusion products, but also to directly target DCs in vivo. This intense experimental effort has culminated in 2010 with the approval by the US FDA of a DC-based preparation (sipuleucel-T, Provenge®) for the treatment of asymptomatic or minimally symptomatic metastatic castration-refractory prostate cancer. As an update to the latest Trial Watch dealing with this exciting field of research (October 2012), here we summarize recent advances in DC-based anticancer regimens, covering both high-impact studies that have been published during the last 13 mo and clinical trials that have been launched in the same period to assess the antineoplastic potential of this variant of cellular immunotherapy.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|